WO2009104100A2 - Combinaisons d’anticorps et leurs procédés de fabrication et d’utilisation - Google Patents

Combinaisons d’anticorps et leurs procédés de fabrication et d’utilisation Download PDF

Info

Publication number
WO2009104100A2
WO2009104100A2 PCT/IB2009/005352 IB2009005352W WO2009104100A2 WO 2009104100 A2 WO2009104100 A2 WO 2009104100A2 IB 2009005352 W IB2009005352 W IB 2009005352W WO 2009104100 A2 WO2009104100 A2 WO 2009104100A2
Authority
WO
WIPO (PCT)
Prior art keywords
target
deposit
cancer
antibody
tumor
Prior art date
Application number
PCT/IB2009/005352
Other languages
English (en)
Other versions
WO2009104100A3 (fr
Inventor
Heinz Peter Vollmers
Frank Hensel
Barbara Power
Leodevico L. Ilag
Arndt-Rene Kelter
Frank Schoenen
Hans-Konrad Mueller-Hermelink
Andreas Thalheimer
Lishanthi Udabage
Stephanie UTE BRÄNDLEIN
Original Assignee
Patrys Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US12/339,737 external-priority patent/US20090202570A1/en
Priority claimed from US12/349,390 external-priority patent/US20090258020A1/en
Priority claimed from US12/350,072 external-priority patent/US20090291083A1/en
Application filed by Patrys Limited filed Critical Patrys Limited
Publication of WO2009104100A2 publication Critical patent/WO2009104100A2/fr
Publication of WO2009104100A3 publication Critical patent/WO2009104100A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2857Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against nuclear receptors, e.g. retinoic acid receptor [RAR], RXR, orphan receptor
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype

Definitions

  • the invention relates to antibody combination compositions, methods of using two or more antibodies alone or ina combination.
  • a combination can include two or more IgMs, two or more IgGs, or an IgM and an IgG.
  • Certain antibodies bind to different types of neoplasia, cancer, tumor and metastasis.
  • Certain antibodies inhibit growth or proliferation of various types of cancer cells and stimulates or induces apoptosis of various types of cancer cells.
  • a series of human monoclonal antibodies isolated from cancer patients, which bind to different tumor-specific surface receptors and induce apoptosis in vitro and in vivo were studied.
  • the antibodies were applied in different combinations to pancreas carcinoma cells and analyzed in cytotoxic assays. Depending on the target, some combinations showed a significant additive or synergistic killing effect, as compared to mono-therapy.
  • a combinatorial treatment with chemotherapy on pancreas and colon cancer cells was also studied. The results indicate that a pretreatment with 5-E 7 U sensitizes cancer cells to antibody treatment.
  • antibody immunotherapies can include cocktails of monoclonal antibodies as well as treatment in combination with other chemotherapeutic protocols.
  • the invention therefore provides combinations of isolated or purified antibodies and functional fragments, and methods of using antibodies and functional fragments in combination (e.g., each antibody administered individually to a subject sequentially or administered to a subject as a combination composition).
  • Antibodes include IgMs and IgGs.
  • IgMs include combinations of IgMs (e.g., two or more IgMs).
  • IgGs include combinations of IgGs (e.g., two or more IgGs).
  • Combinations include combinations of IgMs (e.g., two or more IgMs), combinations of IgGs (e.g., two or more IgGs) and combinations of IgM and IgG.
  • Methods include administering combinations of IgMs (e.g., two or more IgMs), combinations of IgGs (e.g., two or more IgGs) and combinations of IgM and IgG to a subject. Methods further include administering a first IgM to a subject followed by administering a second IgM to a subject; administering a first IgG to a subject followed by administering a second IgG to a subject; administering a first IgM to a subject followed by administering a second IgG to a subject; and administering a first IgG to a subject followed by administering a second IgM to a subject.
  • Exemplary antibodies include LM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2623, deposited on November 6, 2003; CM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC 2584, deposited on March 5, 2003; SAM-6 antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC 2903; PM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2599, deposited on July 2, 2003; PM-2 antibody produced by a hybridoma deposit, such as DSMZ Deposit No.
  • DSM ACC 2600 deposited on July 2, 2003
  • CM-2 antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2598, deposited on July 2, 2003
  • Norm-1 antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2624, deposited on November 6, 2003
  • Norm-2 antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2626, deposited on November 6, 2003
  • BARB3 antibody produced by a hybridoma deposit such as DSMZ Deposit No. DSM ACC2859, deposited on October 10, 2007
  • BARB4 antibody produced by a hybridoma deposit such as DSMZ Deposit No. DSM ACC2876, deposited on December 19, 2007.
  • Exemplary antibodies further include antibody and functional fragments having a heavy and light chain sequence set forth herein, for example, any of SEQ YD NOs: 1 to 58.
  • LM-I binds to LM- 1 target.
  • LM-I target has sequence identity with non-pou domain-containing octamer-binding protein (NONO), also known as 54 kDa nuclear RNA- and DNA-binding protein (p54nrb) and 55 kDa nuclear protein (nmt55).
  • NONO non-pou domain-containing octamer-binding protein
  • p54nrb 54 kDa nuclear RNA- and DNA-binding protein
  • nmt55 nuclear protein
  • LM-I target has a molecular weight of about 54-55 KDa, as determined by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE).
  • NONO/nmt55 can be target for treatment of a neoplasia, cancer, tumor or metastasis.).
  • LM-I antibody or functional fragment thereof binds to one or more of a colorectal cancer, ovarian carcinoma, squamous cell lung carcinoma, small cell lung carcinoma, lobular or ductal mammary carcinoma, melanoma, breast cancer, lung cancer, such as a lung adenocarcinoma, a gastric cancer, a pancreatic cancer, such as a pancreatic adenocarcinoma, a glioma, a sarcoma, a gastrointestinal cancer, a brain tumor, an esophageal cancer, such as an esophagial squamous cell carcinoma, a stomach cancer, an osteosarcoma, a fibrosarcoma, a urinary bladder cancer, a prostate cancer, such as prostate adenocarcinomas, renal cancer, ovarian cancer, testicular cancer, endometrial cancer, cervical cancer, uterine adenocarcinoma, Hodgkin's disease,
  • LM-I antibody or functional fragment thereof also binds to one or more of a lung adenocarinoma cell line Colo-699 (DSMZ accession number ACC 196), lung adenocarinoma cell line DV-90 (DSMZ accession number ACC 307), epidermoid lung carcinoma cell line EPLC-272H (DSMZ accession number ACC 383), and a lung squamous cell carcinoma cell line LOU-NH91 (DSMZ accession number ACC 393).
  • LM-I antibody or functional fragment thereof inhbits or reduces proliferation, or stimulates or induces apoptosis, of one or more of a colorectal cancer, ovarian carcinoma, squamous cell lung carcinoma, small cell lung carcinoma, lobular or ductal mammary carcinoma, melanoma, breast cancer, lung cancer, such as a lung adenocarcinoma, a gastric cancer, a pancreatic cancer, such as a pancreatic adenocarcinoma, a glioma, a sarcoma, a gastrointestinal cancer, a brain tumor, an esophageal cancer, such as an esophagial squamous cell carcinoma, a stomach cancer, an osteosarcoma, a fibrosarcoma, a urinary bladder cancer, a prostate cancer, such as prostate adenocarcinomas, renal cancer, ovarian cancer, testicular cancer, endometrial cancer, cervical cancer, uterine
  • LM-I antibody or functional fragment thereof inhbits or reduces proliferation, or stimulates or induces apoptosis, of one or more of a lung adenocarinoma cell line Colo-699 (DSMZ accession number ACC 196), lung adenocarinoma cell line DV-90 (DSMZ accession number ACC 307), epidermoid lung carcinoma cell line EPLC- 272H (DSMZ accession number ACC 383), and a lung squamous cell carcinoma cell line LOU-NH91 (DSMZ accession number ACC 393).
  • CM-I antibody or functional fragment thereof binds to one or more of a colorectal adenocarcinoma, ovarian cancer, squamous cell lung carcinoma, lobular mammary carcinoma, stomach carcinoma, esophagial squamous cell carcinoma, pancreatic adenocarcinoma, lung adenocarcinoma, ductal mammary carcinoma, uterine adenocarcinoma, or prostate adenocarcinoma cell.
  • CM-I antibody or functional fragment thereof also binds to one or more of HT-29 (ATCC Accession No. HTB-38; DSMZ Accession No. ACC 299), CACO- 2 (ATCC Accession No.
  • CM-I antibody or functional fragment thereof inhbits or reduces proliferation, or stimulates or induces apoptosis, of one or more of a colorectal adenocarcinoma, ovarian cancer, squamous cell lung carcinoma, lobular mammary carcinoma, stomach carcinoma, esophagial squamous cell carcinoma, pancreatic adenocarcinoma, lung adenocarcinoma, ductal mammary carcinoma, uterine adenocarcinoma, prostate adenocarcinoma, or HT-29 (ATCC Accession No. HTB-38; DSMZ Accession No. ACC 299), CACO-2 (ATCC Accession No.
  • SAM-6 binds to SAM-6 targets.
  • An exemplary SAM-6 target has an apparent molecular weight in a range of about 80- 82 kilodaltons (KDa) as determined by denaturing gel electrophoresis.
  • Another exemplary SAM-6 target is apolipoprotein BlOO (apoB lOO), which has a C- terminal LDL receptor binding region.
  • Additional SAM-6 targets include deglycosylated Grp78, LDL (e.g., oxLDL), VLDL, and deglycosylated LDL.
  • SAM-6 antibody can specifically bind to one or more of SAM-6 target, Grp78, deglycosylated Grp78, apoBlOO, LDL (e.g., oxLDL), VLDL, or deglycosylated LDL.
  • LDL e.g., oxLDL
  • VLDL e.g., VLDL
  • deglycosylated LDL e.g., VLDL
  • SAM-6 antibody or functional fragment thereof also binds to one or more of a lung cancer, such as a lung adenocarcinoma, squamous cell lung carcinoma, a gastric cancer such as intestinal type gastric carcinoma, diffuse type gastric carcinoma, colon cancer such as adenocarcinoma of colon, prostate cancer such as a prostate adinocarcinoma, esophagus cancer such as squamous cell carcinoma of esophagus or adenocarcinoma of esophagus, breast cancer such as lobular carcinoma of breast or ductal casrcinoma of breast, prostate cancer such as adenocarcinoma of prostate, ovarian or uterine cancer such as adenocarcinoma of ovary or adenocarcinoma of a uterus cell.
  • a lung cancer such as a lung adenocarcinoma, squamous cell lung carcinoma, a gastric cancer such as intestinal type gastric carcinoma, diffuse
  • SAM-6 antibody or functional fragment thereof also binds to one or more of BXPC-3 (ATCC Deposit No. CRL- 1687), 23132/87 (DSMZ Deposit No. ACC 201) COLO-206F (DSMZ Accession No. ACC 21), COLO-699 (DSMZ Accession No. ACC 196), or LOU-NH91 (DSMZ Accession No. 393) cells.
  • SAM-6 antibody or functional fragment thereof inhbits or reduces proliferation, or stimulates or induces apoptosis, of one or more of a lung cancer, such as a lung adenocarcinoma, squamous cell lung carcinoma, a gastric cancer such as intestinal type gastric carcinoma, diffuse type gastric carcinoma, colon cancer such as adenocarcinoma of colon, prostate cancer such as a prostate adinocarcinoma, esophagus cancer such as squamous cell carcinoma of esophagus or adenocarcinoma of esophagus, breast cancer such as lobular carcinoma of breast or ductal casrcinonia of breast, prostate cancer such as adenocarcinoma of prostate, ovarian or uterine cancer such as adenocarcinoma of ovary or adenocarcinoma of a uterus cell, or BXPC-3 (ATCC Deposit No.
  • a lung cancer such as a lung aden
  • PM-I antibody or functional fragment thereof binds to one or more of a stomach adenocarcinoma, colorectal adenocarcinoma, squamous cell lung carcinoma, lung adenocarcinoma, squamous cell carcinoma of the esophagus, adenocarcinoma of the pancreas, urothel carcinoma of the urinary bladder, renal cell carcinoma of the kidney, adenocarcinoma of the prostate, ductal carcinoma of the breast, lobular carcinoma of the breast, adenocarcinoma of the ovary, adenocarcinoma of the endometrium, or adenocarcinoma of a uterus cell.
  • PM-I antibody or functional fragment thereof also binds to one or more of ASPC- 1 (ATCC Accession No. CRL- 1682) and BXPC-3 (ATCC Accession No. CRL- 1687) cells.
  • PM-I antibody or functional fragment thereof inhbits or reduces proliferation, or stimulates or induces apoptosis, of one or more of a stomach adenocarcinoma, colorectal adenocarcinoma, squamous cell lung carcinoma, lung adenocarcinoma, squamous cell carcinoma of the esophagus, adenocarcinoma of the pancreas, urothel carcinoma of the urinary bladder, renal cell carcinoma of the kidney, adenocarcinoma of the prostate, ductal carcinoma of the breast, lobular carcinoma of the breast, adenocarcinoma of the ovary, adenocarcinoma of the endometrium, adenocarcinoma of a uterus cell, or one or more of ASPC-I (ATCC Accession No. CRL-1682) and BXPC-3 (ATCC Accession No. CRL-1687) cells.
  • ASPC-I ATCC Accession No.
  • PM-2 antibody or functional fragment thereof binds to one or more of a stomach adenocarcinoma, colorectal adenocarcinoma, squamous cell lung carcinoma, lung adenocarcinoma, squamous cell carcinoma of the esophagus, adenocarcinoma of the pancreas, urothel carcinoma of the urinary bladder, renal cell carcinoma of the kidney, adenocarcinoma of the prostate, ductal carcinoma of the breast, lobular carcinoma of the breast, adenocarcinoma of the ovary, or adenocarcinoma of a uterus cell.
  • PM-2 antibody or functional fragment thereof also binds to one or more of HT-29 (ATCC. Accession No. HTB-38; DSMZ Accession No. ACC 299), CACO-2 (ATCC Accession No. HBT-37; DSMZ Accession No. ACC 169), COLO-320 (DSMZ Accession No. ACC 144), COLD- 206F (DSMZ Accession No. ACC 21), ASPC-I (ATCC Accession No. CRL- 1682), or BXPC-3 (ATCC Accession No. CRL- 1687) cells.
  • HT-29 ATCC. Accession No. HTB-38; DSMZ Accession No. ACC 299
  • CACO-2 ATCC Accession No. HBT-37
  • DSMZ Accession No. ACC 169 COLO-320
  • COLD- 206F DSMZ Accession No. ACC 21
  • ASPC-I ATCC Accession No. CRL- 1682
  • BXPC-3 ATCC Accession No. CRL
  • PM-2 antibody or functional fragment thereof inhbits or reduces proliferation, or stimulates or induces apoptosis of one or more of a stomach adenocarcinoma, colorectal adenocarcinoma, squamous cell lung carcinoma, lung adenocarcinoma, squamous cell carcinoma of the esophagus, adenocarcinoma of the pancreas, urothel carcinoma of the urinary bladder, renal cell carcinoma of the kidney, adenocarcinoma of the prostate, ductal carcinoma of the breast, lobular carcinoma of the breast, adenocarcinoma of the ovary, adenocarcinoma of a uterus cell, or HT-29 (ATCC.
  • CM-2 antibody or functional fragment thereof binds to one or more of a colorectal adenocarcinoma or adenocarcinoma of the endometrium cell.
  • CM-2 antibody or functional fragment thereof also binds to one or more of CACO-2 (ATCC Accession No. HBT-37; DSMZ Accession No. ACC 169) or COLO-206F (DSMZ Accession No. ACC 21) cells.
  • CM-2 antibody or functional fragment thereof inhbits or reduces proliferation, or stimulates or induces apoptosis, of one or more of a colorectal adenocarcinoma or adenocarcinoma of the endometrium cell, or CACO-2 (ATCC Accession No. HBT-37; DSMZ Accession No. ACC 169) or COLO-206F (DSMZ Accession No. ACC 21) cells.
  • Norm- 1 antibody or functional fragment thereof binds to one or more of an adenocarcinoma of the colon, a diffuse-type stomach carcinoma, an adenocarcinoma of the pancreas, or an adenocarcinoma of the lung.
  • No ⁇ n- 1 antibody or functional fragment thereof also binds to one or more of EPLC-272H (DSMZ (Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH; German Collection of Microorganisms and Cell Cultures) Accession Number ACC 383), Colo-699 (DSMZ Accession Number ACC 196), CACO-2 (DSMZ Accession Number ACC 169, ATCC (American Type Culture Collection) Accession Number HTB-37), Colo-206F (DSMZ Accession Number ACC 21), 23132/87 (DSMZ Accession Number ACC 201), ASPC-I (ATCC Accession Number CRL-1682), DU-145 (DSMZ Accession Number ACC 261, ATCC Accession Number HTB-81), or BM1604 (DSMZ Accession Number ACC 298) cells.
  • DSMZ Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH; German Collection of Microorganisms and Cell Cultures
  • CACO-2 DMZ Accession Number ACC 169, ATCC (American Type Culture Collection)
  • Norm-1 antibody or functional fragment thereof inhbits or reduces proliferation, or stimulates or induces apoptosis of one or more of an adenocarcinoma of the colon, a diffuse-type stomach carcinoma, an adenocarcinoma of the pancreas, an adenocarcinoma of the lung, or EPLC-272H (DSMZ (Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH; German Collection of Microorganisms and Cell Cultures) Accession Number ACC 383), Colo-699 (DSMZ Accession Number ACC 196), CACO-2 (DSMZ Accession Number ACC 169, ATCC (American Type Culture Collection) Accession Number HTB-37), Colo-206F (DSMZ Accession Number ACC 21), 23132/87 (DSMZ Accession Number ACC 201), ASPC-I (ATCC Accession Number CRL-1682), DU-145 (DSMZ Accession Number ACC 261, ATCC Accession Number HTB-81), or BM
  • Norm-2 antibody or functional fragment thereof binds to one or more of an adenocarcinoma of the colon, a diffuse-type stomach carcinoma, an adenocarcinoma of the pancreas, or an adenocarcinoma of the lung.
  • Norm-1 antibody or functional fragment thereof also binds to one or more of EPLC-272H (DSMZ (Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH; German Collection of Microorganisms and Cell Cultures) Accession Number ACC 383), Colo-699 (DSMZ Accession Number ACC 196), CACO-2 (DSMZ Accession Number ACC 169, ATCC (American Type Culture Collection) Accession Number HTB-37), Colo-206F (DSMZ Accession Number ACC 21), 23132/87 (DSMZ Accession Number ACC 201), ASPC-I (ATCC Accession Number CRL-1682), DU-145 (DSMZ Accession Number ACC 261, ATCC Accession Number HTB-81), or BM1604 (DSMZ Accession Number ACC 298) cells.
  • DSMZ Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH; German Collection of Microorganisms and Cell Cultures
  • CACO-2 DMZ Accession Number ACC 169, ATCC (American Type Culture Collection) Accession
  • Norm-2 antibody or functional fragment thereof inhbits or reduces proliferation, or stimulates or induces apoptosis of one or more of an adenocarcinoma of the colon, a diffuse-type stomach carcinoma, an adenocarcinoma of the pancreas, an adenocarcinoma of the lung, or EPLC-272H (DSMZ (Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH; German Collection of Microorganisms and Cell Cultures) Accession Number ACC 383), Colo-699 (DSMZ Accession Number ACC 196), CACO-2 (DSMZ Accession Number ACC169, ATCC (American Type Culture Collection) Accession Number HTB-37), Colo-206F (DSMZ Accession Number ACC 21), 23132/87 (DSMZ Accession Number ACC 201), ASPC-I (ATCC Accession Number CRL-1682), DU-145 (DSMZ Accession Number ACC 261, ATCC Accession Number HTB-81), or BM160
  • BARB3 antibody binds to one or more of a human adenocarcinoma, human squamous cell carcinoma, human carcinoid carcinoma, human invasive ductal carcinoma, or human germ cell carcinoma of any of stomach, lung, colon, pancreas, esophagus, prostate, breast or testis.
  • BARB3 antibody or functional fragment thereof also binds to one or more of a stomach adenocarcinoma cell, a lung adenocarcinoma cell, a pancreas adenocarcinoma cell, a colon adenocarcinoma cell, a breast adenocarcinoma cell, or an esophagus squamous cell carcinoma, a human adenocarcinoma, human squamous cell carcinoma, human carcinoid carcinoma, human invasive ductal carcinoma, or a human germ cell carcinoma in any of stomach, lung, colon, pancreas, esophagus, prostate, breast or testis, or a pancreas cancer cell line BXPC-3 (ATCC Deposit No. CRL-1687) or a stomach cancer cell line 23132/87 (DSMZ Deposit No. ACC 201).
  • BARB 3 antibody or functional fragment thereof inhbits or reduces proliferation, or stimulates or induces apoptosis, of one one or more of a stomach adenocarcinoma cell, a lung adenocarcinoma cell, a pancreas adenocarcinoma cell, a colon adenocarcinoma cell, a breast adenocarcinoma cell, or an esophagus squamous cell carcinoma, a human adenocarcinoma, human squamous cell carcinoma, human carcinoid carcinoma, human invasive ductal carcinoma, or a human germ cell carcinoma in any of stomach, lung, colon, pancreas, esophagus, prostate, breast or testis, or a pancreas cancer cell line BXPC-3 (ATCC Deposit No.
  • BARB4 binds to BARB4 target.
  • BARB4 target has sequence identity with TATA-binding protein-associated factor 15 (TAF 15 polypeptide).
  • BARB4 target has a molecular weight of about 70-85 KDa, as determined by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS- PAGE).
  • BARB4 antibody also binds to one or more of an adenocarcinoma cell or a squamous cell carcinoma, a stomach adenocarcinoma cell, a lung adenocarcinoma cell, a pancreas adenocarcinoma cell, a colon adenocarcinoma cell, a breast adenocarcinoma cell, or an esophagus squamous cell carcinoma.
  • BARB4 antibody or functional fragment thereof also binds to one or more of a human adenocarcinoma, human squamous cell carcinoma, human carcinoid carcinoma, human invasive ductal carcinoma, or human germ cell carcinoma of any of stomach, lung, colon, pancreas, esophagus, prostate, breast or testis.
  • B ARB4 antibody or functional fragment thereof particularly binds to one or more of pancreas cancer cell line BXPC-3 (ATCC Deposit No. CRL- 1687), colon cancer cell line HT-29 (ATCC Deposit No. HTB-38) or stomach cancer cell line 23132/87 (DSMZ Deposit No. ACC 201).
  • BARB4 antibody or functional fragment thereof inhbits or reduces proliferation, or stimulates or induces apoptosis, of one or more of a stomach adenocarcinoma cell, a lung adenocarcinoma cell, a pancreas adenocarcinoma cell, a colon adenocarcinoma cell, a breast adenocarcinoma cell, or an esophagus squamous cell carcinoma, a human adenocarcinoma, human squamous cell carcinoma, human carcinoid carcinoma, human invasive ductal carcinoma, or a human germ cell carcinoma in any of stomach, lung, colon, pancreas, esophagus, prostate, breast or testis, or a pancreas cancer cell line BXPC-3 (ATCC Deposit No. CRL- 1687), a colon cancer cell line HT-29 (ATCC Deposit No. HTB-38) or a stomach cancer cell line 23132/87 (DSM
  • Antibodies and functional fragments include a heavy or light chain variable region sequence with about 60% or more identity to a reference heavy or light chain sequence variable region, as represented by any antibody produced by a hybridoma set forth herein, or any sequence as set forth herein, for example, any of SEQ ID NOs: 1 to 58.
  • an antibody or subsequence thereof includes a sequence at least 60 % or more (e.g., 65%, 70%, 75%, 80%, 85%, 90%, 95%, etc.) identical to a heavy or light chain variable region sequence set forth in any of SEQ ID NOs: 1 to 58, or a sequence at least 60% or more (e.g., 65%, 70%, 75%, 80%, 85%, 90%, 95%, etc.) identical to a heavy or light chain variable region sequence set forth in any of SEQ ID NOs: 1 to 58.
  • an antibody or subsequence includes a sequence at least 60 % or more (e.g., 65%, 70%, 75%, 80%, 85%, 90%, 95%, etc.) identical to a heavy or light chain variable region sequence in any of SEQ ID NOs: 1 to 58, and a sequence at least 60% or more (e.g., 65%, 70%, 75%, 80%, 85%, 90%, 95%, etc.) identical to a heavy or light chain variable region sequence in any of SEQ ID NOs: 1 to 58.
  • an antibody or subsequence includes a sequence at least 80-85%, 85-90%, 90-95%, or 95-100% identical to one or more CDRs m a heavy or light chain variable region sequence set forth in any of SEQ ID NOs.1 to 58.
  • Antibodies and functional fragments further include those that have one or more amino acid additions, deletions or substitutions of a heavy or light chain variable region sequences disclosed herein, such as in any of SEQ ID NOs: 1 to 58, or a heavy or light chain sequence produced by a hyb ⁇ doma deposit, such as DSMZ Deposit No. DSM ACC2623, deposited on November 6, 2003; a hybndoma deposit, such as DSMZ Deposit No. DSM ACC 2584, deposited on March 5. 2003; a hybndoma deposit, such as DSMZ Deposit No. DSM ACC2599, deposited on July 2, 2003; a hybndoma deposit, such as DSMZ Deposit No.
  • DSM ACC 2600 deposited on July 2, 2003; a hyb ⁇ doma deposit, such as DSMZ Deposit No. DSM ACC2598, deposited on July 2, 2003, a hybndoma deposit, such as DSMZ Deposit No. DSM ACC2624, deposited on November 6, 2003; a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2626, deposited on November 6, 2003; a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2859, deposited on October 10, 2007; or a hybndoma deposit, such as DSMZ Deposit No. DSM ACC2876, deposited on December 19, 2007.
  • an antibody or functional fragment has a sequence at least 80-85%, 85-90%, 90-95%, or 95-100% identical to a heavy cham variable region sequence, or has a sequence at least 80-85%, 85-90%, 90- 95%, or 95-100% identical to a light chain variable region sequence, of LM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2623, deposited on November 6, 2003; CM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC 2584, deposited on March 5, 2003; SAM-6 antibody produced by a hybridoma deposit, such as DSMZ Deposit No.
  • an antibody or functional fragment has a heavy or light chain sequence with 100% identity to one or more CDRs in a heavy or light chain variable region sequence as represented by any antibody of LM- 1 antibody produced by a hybridoma deposit, such as DSMZ Deposit No.
  • DSM ACC2623 deposited on November 6, 2003; CM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC 2584, deposited on March 5, 2003; SAM-6 antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC 2903; PM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2599, deposited on July 2, 2003; PM-2 antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC 2600, deposited on July 2, 2003; CM-2 antibody produced by a hybridoma deposit, such as DSMZ Deposit No.
  • DSM ACC2876 deposited on December 19, 2007, or any heavy or light chain variable region sequence set forth herein, such as any of SEQ K) NOs: 1 to 58, and has less than 100% identity to a region outside of the CDRs in the heavy or light chain variable region sequence.
  • Antibodies and functional fragments thereof can have a binding affinity within about 1-5000 fold of the binding affinity of a reference antibody, e.g., as represented by LM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2623, deposited on November 6, 2003; CM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC 2584, deposited on March 5, 2003; SAM-6 antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC 2903; PM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No.
  • DSM ACC2859 deposited on October 10, 2007; or BARB4 antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2876, deposited on December 19, 2007, or any of SEQ ID NO:1 to 58, for binding to an antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated Grp78, apoB lOO, LDL (e.g., oxLDL), VLDL, or deglycosylated LDL, Barb4 target, TAF- 15 polypeptide), or compete for binding to a cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/n
  • antibodies and functional fragments have a binding affinity within about 1-5000 fold of the binding affinity of a reference antibody (as represented by LM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2623, deposited on November 6, 2003; CM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC 2584, deposited on March 5, 2003; SAM-6 antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC 2903; PM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No.
  • LM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2623, deposited on November 6, 2003
  • CM-I antibody produced by a hybridoma deposit such as DSMZ Deposit No. DSM ACC 2584, deposited on March 5, 2003
  • SAM-6 antibody produced by a hybridoma deposit, such as DSMZ Deposit No. D
  • DSM ACC2859 deposited on October 10, 2007; or BARB4 antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2876, deposited on December 19, 2007, or heavy and light chain sequences set forth as any of SEQ ED NO:1 to 58 for binding to any cell or antigen cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated Grp78, apoBlOO, LDL (e.g., oxLDL), VLDL, or deglycosylated LDL, Barb4 target, TAF- 15 polypeptide), or compete for binding to a cell or antigen (e.g., CM- 1 target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target,
  • an antibody or functional fragment has a binding affinity within about KD 10 "5 M to about KD 10 "13 M for binding to one or more cells or cell lines set forth herein (e.g., an adenocarcinoma cell, a squamous cell carcinoma, a stomach adenocarcinoma cell, a lung adenocarcinoma cell, a pancreas adenocarcinoma cell, a colon adenocarcinoma cell, a breast adenocarcinoma cell, an esophagus squamous cell carcinoma, etc.).
  • an adenocarcinoma cell e.g., an adenocarcinoma cell, a squamous cell carcinoma, a stomach adenocarcinoma cell, a lung adenocarcinoma cell, a pancreas adenocarcinoma cell, a colon adenocarcinoma cell, a breast adeno
  • Antibodies of the invention include IgG, IgA, IgM, IgE and IgD.
  • an IgG is an IgGl, IgG2, IgG3, or IgG4.
  • Antibody functional fragments and subsequences include functional fragments and subsequences of the various antibodies set forth herein.
  • a functional fragment or a subsequence is an Fab, Fab', F(ab'> 2 , Fv, Fd, single-chain Fv (scFv), disulfide-linked Fvs (sdFv), V L , V H , trispecific (Fab3), bispecific (Fab 2 ), diabody ((V L -V H ) 2 or (V H -V L ) 2 ), triabody (trivalent), tetrabody (tetravalent), minibody ((SCF V -C H 3) 2 ), bispecific single-chain Fv (Bis-scFv), IgGdeltaCH2, scFv-Fc or (scFv) 2 -Fc.
  • a functional fragment or a subsequence of a full length antibody heavy or light chain, or a heavy or light chain variable region has a length from about 20-30, 30- 50, 50-100, 100-150, 150-200, 200-250, 250-300, 300-400, 400-500, amino acid residues.
  • Antibodies and subsequences can include a heterologous domain.
  • a heterologous domain includes a detectable label, tag or cytotoxic agent.
  • a detectable label or tag is an enzyme, enzyme substrate, ligand, receptor, radionuclide, a T7-, His-, myc-, HA- or FLAG-tag, electron-dense reagent, energy transfer molecule, paramagnetic label, fluorophore, chromophore, chemi-luminescent agent, or a bio-luminescent agent.
  • Isolated and purified cells as well as transformed host cells can express an antibody or subsequence thereof that includes a sequence at least 60% or more (e.g., 65%, 70%, 75%, 80%, 85%, 90%, 95%, etc.) identical to a heavy chain variable region sequence produced by any anbtibody of LM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2623, deposited on November 6, 2003; CM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC 2584, deposited on March 5, 2003; SAM-6 antibody produced by a hybridoma deposit, such as DSMZ Deposit No.
  • a hybridoma deposit such as DSMZ Deposit No.
  • Such cells include eukaryotic and non-eukaryotic cells, which can stably or transiently express antibody or subsequence thereof, or be stably or transiently transformed with the nucleic acid or vector that encodes antibody or subsequence thereof.
  • Kits can include an antibody or functional fragment thereof, hi particular aspects, a kit includes an antibody or functional fragment thereof with a heavy or light chain variable region sequence of LM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2623, deposited on November 6, 2003; CM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC 2584, deposited on March 5, 2003; SAM-6 antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC 2903; PM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No.
  • a hybridoma deposit such as DSMZ Deposit No.
  • Kits further include antibodies and functional fragments that include a heavy or light chain variable region sequence with about 60% (e.g., 65%, 70%, 75%, 80%, 85%, 90%, 95%, etc.) or more identity to a heavy or light chain sequence variable regions of a reference antibody or functional fragment.
  • antibody or functional fragment includes a heavy or light chain sequence variable region of LM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2623, deposited on November 6, 2003; CM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC 2584, deposited on March 5, 2003; SAM-6 antibody produced by a hybridoma deposit, such as DSMZ Deposit No.
  • DSM ACC2626 deposited on November 6, 2003; BARB3 antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2859, deposited on October 10, 2007; or BARB4 antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2876, deposited on December 19, 2007,or any of SEQ ID NO: l to 58.
  • kits includes an anti-cell proliferative or immune enhancing treatment or therapeutic agent, or an antineoplastic, anti-cancer or anti-tumor agent, or an article of manufacture (e.g., for delivering the antibody, anti-cell proliferative or immune enhancing treatment or therapy into a subject locally, regionally or systemically).
  • the instructions are for treating undesirable cell proliferation or a cell proliferative disorder (e.g., a neoplasia, tumor cancer or metastasis.
  • compositions include an antibody or functional fragment and a pharmaceutically acceptable carrier or excipient.
  • a composition includes two or more antibody or functional fragments.
  • an antibody is designated LM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2623, deposited on November 6, 2003; CM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC 2584, deposited on March 5, 2003; SAM-6 antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC 2903; PM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No.
  • DSM ACC2859 deposited on October 10, 2007; or BARB4 antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2876, deposited on December 19, 2007, or comprises a heavy and light chain sequence of any of SEQ ID NO:1 to 58, and a pharmaceutically acceptable carrier or excipient.
  • Methods include administering an antibody or functional fragment, such as administering an antibody or functional fragment and a second therapeutic agent (e.g., a chemotheoprapeutic agent) in combination to a subject
  • a second therapeutic agent e.g., a chemotheoprapeutic agent
  • Such methods therefore include simultaneous administration of an antibody or functional fragment and a second therapeutic agent, as well as administering a composition that includes an antibody or functional fragment and a second therapeutic agent.
  • Methods also mclude administering a first antibody or functional fragment to a subject followed by administering a second therapeutic agent.
  • Methods moreover include administering an antibody or functional fragment, such as administering two antibodies or functional fragments m combination to a subject. Such methods therefore mclude simultaneous administration of two or more antibodies or functional fragments, or administering a composition that includes two or more antibodies. Methods also include administering a first antibody or functional fragment to a subject followed by administering a second antibody or functional fragment.
  • Antibodies, functional fragments and modified forms aie useful for treating a subject in need of treatment The invention therefore provides methods of using antibodies and functional fragments in treatment (e.g., therapeutic or prophylactic) of a subject having or at risk of having undesirable cell proliferation, such as a cell proliferative or hyperprohferative disorder.
  • a method includes administering two antibodies or functional fragments in combination to a subject having or at ⁇ sk of having undesirable cell proliferation (e.g., a cell proliferative disorder) an amount effective to treat undesirable cell proliferation.
  • a method in another embodiment, includes administering a first antibody or functional fragment to a subject followed by admmistermg a second antibody or functional fragment to a subject having or at risk of having undesirable cell proliferation (e.g., a cell proliferative disorder) an amount effective to treat undesirable cell proliferation.
  • a cell proliferative disorder is a metastatic or non-metastatic, solid or liquid neoplasia, malignancy, tumor or cancer.
  • undesirable cell proliferation affects or is present at least in part in brain, head or neck, breast, esophagus, mouth, nasopharynx, nose or sinuses, stomach, duodenum, ileum, jejunum, lung, liver, pancreas, kidney, adrenal gland, thyroid, bladder, colon, rectum, prostate, uterus, cervix, ovary, bone marrow, lymph, blood, bone, testes, skin or muscle, or hematopoetic system.
  • undesirable cell proliferation includes a neoplasia, tumor, cancer or metastasis that affects or is at least in part present in breast, lung, thyroid, head and neck, nasopharynx, nose or sinuses, brain, spine, adrenal gland, thyroid, lymph, gastrointestinal tract, mouth, esophagus, stomach, duodenum, ileum, jejunum, small intestine, colon, rectum, genito-urinary tract, uterus, ovary, cervix, bladder, testicle, penis, prostate, kidney, pancreas, adrenal gland, liver, bone, bone marrow, lymph, blood, muscle, skin or is hematopoetic.
  • a cell proliferative disorder includes a neoplasia, tumor, cancer or metastasis that affects or is at least in part present in breast, lung, thyroid, head and neck, nasopharynx, nose or sinuses, brain, spine, adrenal gland, thyroid, lymph,
  • a neoplasia, tumor, cancer or metastasis is a sarcoma, carcinoma, adenocarcinoma, melanoma, myeloma, blastoma, glioma, lymphoma leukemia.
  • a neoplasia, tumor or cancer is a lung adenocarcinoma, lung carcinoma, diffuse or interstitial gastric carcinoma, colon adenocarcinoma, prostate adenocarcinoma, esophagus carcinoma, breast carcinoma, pancreas adenocarcinoma, ovarian adenocarcinoma, or uterine adenocarcinoma, or a metastasis thereof.
  • Methods also include treating a subject having or at risk of having a metastasis.
  • a method includes administering an amount effective to reduce or inhibit spread or dissemination of a tumor, cancer or neoplasia to other sites, locations or regions within the subject.
  • a method reduces or inhibits metastasis of a primary tumor or cancer to one or more other sites, the formation or establishment of a metastasis at one or more other sites, thereby inhibiting or reducing tumor or cancer relapse or tumor or cancer progression.
  • a method reduces or inhibits growth, proliferation, mobility or invasiveness of tumor or cancer cells that potentially or do develop metastases (e.g, disseminated tumor cells); reduces or inhibits formation or establishment of metastases arising from a primary tumor or cancer to one or more other sites, locations or regions distinct from the primary tumor or cancer; reduces or inhibits growth or proliferation of a metastasis at one or more other sites, locations or regions distinct from the primary tumor or cancer after the metastasis has formed or has been established; or reduces or inhibits formation or establishment of additional metastasis after the metastasis has been formed or established.
  • metastases e.g, disseminated tumor cells
  • a neoplasia, tumor or cancer, or metastasis is progressively worsening or is in remission.
  • treatment results in alleviating or ameliorating one or more adverse physical symptoms associated with a cell proliferative disorder, or a neoplasia, tumor or cancer, or reduces or decreases neoplasia, tumor or cancer volume, inhibits or prevents an increase in neoplasia, tumor or cancer volume, inhibits neoplasia, tumor or cancer progression or worsening, stimulates neoplasia, tumor or cancer cell lysis or apoptosis, or inhibits, reduces or decreases neoplasia, tumor or cancer proliferation or metastasis, or prolongs or extends lifespan of the subject, or improves the quality of life of the subject.
  • Methods include administration to a subject locally, regionally, or systemically.
  • exemplary subjects e.g., mammals such as humans
  • exemplary subjects include candidates for, and those undergoing, or having undergone an anti-cell proliferative or anti-hyperproliferative disorder (e.g., anti-neoplastic, anti-tumor, anti-cancer or anti-metastasis) or immune-enhancing treatment or therapy.
  • an anti-cell proliferative or anti-hyperproliferative disorder e.g., anti-neoplastic, anti-tumor, anti-cancer or anti-metastasis
  • immune-enhancing treatment or therapy e.g., anti-neoplastic, anti-tumor, anti-cancer or anti-metastasis
  • a method includes administering to a subject an antibody or functional fragment (e.g., with a heavy or light chain variable region of any sequence set forth herein, such as any of SEQ ID NOs: 1 to 58) as a combination with another antibody or second therapeutic agent, or adminstering the antibody or functional fragment and the other antibody or functional fragment or second therapeutic agent sequentially (e.g., an anti-cell proliferative or immune-enhancing treatment or therapy to a subject (e.g., prior to, substantially contemporaneously with or following each other).
  • an antibody or functional fragment e.g., with a heavy or light chain variable region of any sequence set forth herein, such as any of SEQ ID NOs: 1 to 58
  • an anti-cell proliferative or immune-enhancing treatment or therapy includes surgical resection, radiotherapy, radiation therapy, chemotherapy, immunotherapy, hyperthermia, an alkylating agent, antimetabolite, plant extract, plant alkaloid, nitrosourea, hormone, nucleoside or nucleotide analogue, a lymphocyte, plasma cell, macrophage, dendritic cell, NK cell or B -cell, an antibody, a cell growth factor, a cell survival factor, a cell differentiative factor, a cytokine or a chemokine.
  • Figures IA -IB BXPC-3 Trypan Blue Viability with antibodies alone, or in combination.
  • the combinations are denoted “cocktail” and immediately follow the respective antibodies individually: A) PM-2 and Norm-2; CM-I and PM-I; and B) SAM-6 and LM-I; and SAM-5 and PM-I.
  • Figure 2 MTT Assay on BXPC-3 cells with Norm-2 antibody alone and in combination with 5-FU.
  • Figure 3 FACS Analysis with NORM-2 on 5-FU sensitized BxPC-3 and HT29 cells.
  • the invention is based, at least in part, on antibodies that bind to various neoplastic, cancer, tumor and metastatic cells, and combining the antibodies with second therapeutic agents, or therapeutic methods.
  • Such combinations include combination compositions as well as combining antibodies with other methods of treatment.
  • antibodies can be physically mixed or combined with a second therapeutic agent, for example another antibody or another (non-antibody) agent such as a chemotherapeutic agent.
  • antibodies can but need not be be physically mixed or combined with a second therapeutic agent, such as another antibody or another (non- antibody) agent such as a chemotherapeutic agent.
  • an antibody be administered prior to or after a second therapeutic agent, such as another antibody or another (non-antibody) agent such as a chemotherapeutic agent is administered.
  • a second therapeutic agent such as another antibody or another (non-antibody) agent such as a chemotherapeutic agent is administered.
  • the timing of administration relative to a second therapeutic agent can be within seconds (1-10, 20-30, 30-45, 45-60) minutes (1- 10, 20-30, 30-45, 45-60), hours (1-12, 12-24, 24-48, 48-72), days (1-10, 20-30. 30-45, 45-60) or weeks (1-4, 4-8, 8-12, 12-24) prior to or after a second therapeutic agent is administered.
  • Non-limiting exemplary antibodies and functional fragments include LM- 1 antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2623, deposited on November 6, 2003; CM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC 2584, deposited on March 5, 2003; SAM-6 antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC 2903; PM-I antibody produced by a hybridoma deposit, such as DSMZ Deposit No. DSM ACC2599, deposited on July 2, 2003; PM-2 antibody produced by a hybridoma deposit, such as DSMZ Deposit No.
  • DSM ACC 2600 deposited on July 2, 2003
  • CM-2 antibody produced by a hybridoma deposit such as DSMZ Deposit No. DSM ACC2598, deposited on July 2, 2003
  • Norm-1 antibody produced by a hybridoma deposit such as DSMZ Deposit No. DSM ACC2624, deposited on November 6, 2003
  • Norm-2 antibody produced by a hybridoma deposit such as DSMZ Deposit No. DSM ACC2626, deposited on November 6, 2003
  • BARB4 antibody produced by a hybridoma deposit such as DSMZ Deposit No. DSM ACC2876, deposited on December 19, 2007.
  • Exemplary antibodies and functional fragments further include antibody and functional fragments having any heavy and light chain sequence set forth herein, such as any SEQ ID NOs: 1 to 58.
  • Each of the foregoing antibodies or functional fragments is able to inhibit or reduce proliferation of various neoplastic, cancer, tumor and metastatic cells.
  • Each antibody or functional fragment is also able to stimulate or induce apoptosis of various neoplastic, cancer, tumor and metastatic cells.
  • Non-limiting examples for LM- 1 antibody or functional fragment thereof include one or more of a colorectal cancer, ovarian carcinoma, squamous cell lung carcinoma, small cell lung carcinoma, lobular or ductal mammary carcinoma, melanoma, breast cancer, lung cancer, such as a lung adenocarcinoma, a gastric cancer, a pancreatic cancer, such as a pancreatic adenocarcinoma, a glioma, a sarcoma, a gastrointestinal cancer, a brain tumor, an esophageal cancer, such as an esophagial squamous cell carcinoma, a stomach cancer, an osteosarcoma, a fibrosarcoma, a urinary bladder cancer, a prostate cancer, such as prostate adenocarcinomas, renal cancer, ovarian cancer, testicular cancer, endometrial cancer, cervical cancer, uterine adenocarcinoma, Hodgkin's
  • DSMZ accession number ACC 307 epidermoid lung carcinoma cell line EPLC- 272H (DSMZ accession number ACC 383), or a lung squamous cell carcinoma cell line LOU-NH91 (DSMZ accession number ACC 393).
  • Non- limiting examples for CM-I antibody or functional fragment thereof includecolorectal adenocarcinoma, ovarian cancer, squamous cell lung carcinoma, lobular mammary carcinoma, stomach carcinoma, esophagial squamous cell carcinoma, pancreatic adenocarcinoma, lung adenocarcinoma, ductal mammary carcinoma, uterine adenocarcinoma, or prostate adenocarcinoma cell, HT-29 (ATCC Accession No. HTB-38; DSMZ Accession No. ACC 299), CACO-2 (ATCC Accession No. HBT-37; DSMZ Accession No. ACC 169), COLO-320 (DSMZ Accession No. ACC 144), COLO-206F (DSMZ Accession No. ACC 21), or COLO-678 (DSMZ Accession No. 194) cells.
  • HT-29 ATCC Accession No. HTB-38; DSMZ Accession No. ACC 29
  • Non-limiting examples for SAM-6 antibody or functional fragment thereof include lung adenocarcinoma, squamous cell lung carcinoma, a gastric cancer such as intestinal type gastric carcinoma, diffuse type gastric carcinoma, colon cancer such as adenocarcinoma of colon, prostate cancer such as a prostate adinocarcinoma, esophagus cancer such as squamous cell carcinoma of esophagus or adenocarcinoma of esophagus, breast cancer such as lobular carcinoma of breast or ductal casrcinoma of breast, prostate cancer such as adenocarcinoma of prostate, ovarian or uterine cancer such as adenocarcinoma of ovary or adenocarcinoma of a uterus cell, BXPC-3 (ATCC Deposit No.
  • a gastric cancer such as intestinal type gastric carcinoma, diffuse type gastric carcinoma
  • colon cancer such as adenocarcinoma of colon
  • prostate cancer such as a prostate
  • Non-limiting examples for PM-I antibody or functional fragment thereof include stomach adenocarcinoma, colorectal adenocarcinoma, squamous cell lung carcinoma, lung adenocarcinoma, squamous cell carcinoma of the esophagus, adenocarcinoma of the pancreas, urothel carcinoma of the urinary bladder, renal cell carcinoma of the kidney, adenocarcinoma of the prostate, ductal carcinoma of the breast, lobular carcinoma of the breast, adenocarcinoma of the ovary, adenocarcinoma of the endometrium, adenocarcinoma of a uterus cell, ASPC-I (ATCC Accession No.
  • Non-limiting examples for PM-2 antibody or functional fragment include stomach adenocarcinoma, colorectal adenocarcinoma, squamous cell lung carcinoma, lung adenocarcinoma, squamous cell carcinoma of the esophagus, adenocarcinoma of the pancreas, urothel carcinoma of the urinary bladder, renal cell carcinoma of the kidney, adenocarcinoma of the prostate, ductal carcinoma of the breast, lobular carcinoma of the breast, adenocarcinoma of the ovary, adenocarcinoma of a uterus cell, or HT-29 (ATCC.
  • CM-2 antibody or functional fragment thereof include colorectal adenocarcinoma or adenocarcinoma of the endometrium cell, CACO-2 (ATCC Accession No. HBT-37; DSMZ Accession No. ACC 169) or COLO-206F (DSMZ Accession No. ACC 21 ) cells.
  • Non-limiting examples for Norm-1 antibody or functional fragment include adenocarcinoma of the colon, a diffuse-type stomach carcinoma, an adenocarcinoma of the pancreas, an adenocarcinoma of the lung, EPLC-272H (DSMZ (Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH; German Collection of Microorganisms and Cell Cultures) Accession Number ACC 383), Colo-699 (DSMZ Accession Number ACC 196), CACO-2 (DSMZ Accession Number ACC 169, ATCC (American Type Culture Collection) Accession Number HTB-37), Colo-206F (DSMZ Accession Number ACC 21), 23132/87 (DSMZ Accession Number ACC 201), ASPC-I (ATCC Accession Number CRL-1682), DU-145 (DSMZ Accession Number ACC 261, ATCC Accession Number HTB-81), or BM1604 (DSMZ Accession Number ACC 298) cells.
  • DSMZ Deutsche Sammlung von Mikroorganisme
  • Non-limiting examples for Norm-2 antibody or functional fragment include adenocarcinoma of the colon, a diffuse-type stomach carcinoma, an adenocarcinoma of the pancreas, an adenocarcinoma of the lung, or EPLC- 272H (DSMZ (Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH; German Collection of Microorganisms and Cell Cultures) Accession Number ACC 383), Colo-699 (DSMZ Accession Number ACC 196), CACO-2 (DSMZ Accession Number ACC 169, ATCC (American Type Culture Collection) Accession Number HTB-37), Colo-206F (DSMZ Accession Number ACC 21), 23132/87 (DSMZ Accession Number ACC 201), ASPC-I (ATCC Accession Number CRL-1682), DU-145 (DSMZ Accession Number ACC 261 , ATCC Accession Number HTB-81), or BM1604 (DSMZ Accession Number ACC 298) cells.
  • DSMZ Deutsche
  • Non-limiting examples for BARB3 antibody or functional fragment include human adenocarcinoma, human squamous cell carcinoma, human carcinoid carcinoma, human invasive ductal carcinoma, or human germ cell carcinoma of any of stomach, lung, colon, pancreas, esophagus, prostate, breast or testis, stomach adenocarcinoma cell, a lung adenocarcinoma cell, a pancreas adenocarcinoma cell, a colon adenocarcinoma cell, a breast adenocarcinoma cell, or an esophagus squamous cell carcinoma, a human adenocarcinoma, human squamous cell carcinoma, human carcinoid carcinoma, human invasive ductal carcinoma, or a human germ cell carcinoma in any of stomach, lung, colon, pancreas, esophagus, prostate, breast or testis, or a pancreas cancer cell line BXPC-3 (ATCC Deposit
  • Non-limiting examples for BARB4 antibody or functional fragment include squamous cell carcinoma, a stomach adenocarcinoma cell, a lung adenocarcinoma cell, a pancreas adenocarcinoma cell, a colon adenocarcinoma cell, a breast adenocarcinoma cell, an esophagus squamous cell carcinoma, a human adenocarcinoma, human squamous cell carcinoma, human carcinoid carcinoma, human invasive ductal carcinoma, or human germ cell carcinoma of any of stomach, lung, colon, pancreas, esophagus, prostate, breast or testis, or pancreas cancer cell line BXPC-3 (ATCC Deposit No. CRL- 1687), colon cancer cell line HT-29 (ATCC Deposit No. HTB-38) or stomach cancer cell line 23132/87 (DSMZ Deposit No. ACC 201).
  • BXPC-3 ATCC Deposit No. CRL- 1687
  • Antibodies include polyclonal and monoclonal antibodies. Antibodies are proteins which include amino acids, or “residues,” covalently linked by an amide bond or equivalent.
  • the term “monoclonal,” when used in reference to an antibody refers to an antibody that is based upon, obtained from or derived from a single clone, including any eukaryotic, prokaryotic, or phage clone. A “monoclonal” antibody is therefore defined herein structurally, and not the method by which it is produced.
  • Antibodies of the invention can belong to any antibody class, IgM, IgG, IgE, IgA, IgD, or subclass. Exemplary subclasses for IgG are IgGi, IgG 2 , IgG 3 and IgG 4 .
  • Antibodies of the invention can have kappa or lambda light chain sequences, either full length as in naturally occurring antibodies, mixtures thereof (i.e., fusions of kappa and lambda chain sequences), and subsequences/fragments thereof.
  • Naturally occurring antibody molecules contain two kappa or two lambda light chains. The primary difference between kappa and lambda light chains is in the sequences of the constant region.
  • Predicted CDRs of which there are three in each of heavy and light chain, are conveniently denoted herein as LC-CDRl, LC-CDR2 and LC- CDR3; and HC-CDRl, HC-CDR2 and HC-CDR3.
  • Predicted CDR sequences of LM- 1 heavy chain are CDRl, GGSISSGGYY, CDR2, GY IYYSGSTYYN, and CDR3, RVDARYDYVWGSYR.
  • Predicted CDR sequences of LM- 1 light chain are located at positions 16-22, 37-52 and 85-103, CDRl, SGSSSNIGNNYVS, CDR2, DNNKRPS, and CDR3, GTWDSSLSA.
  • Additional antibodies and fragments include LM-I variant heavy and light chain sequences.
  • Amino acid sequence of LM-I heavy chain variable (VH) region sequence as represented by SEQ ID NO:3 (IBTAl.16VH):
  • VH heavy chain variable region sequence
  • SEQ ID NO:4 (1BTA1.7 VH):
  • VH heavy chain variable region sequence
  • SEQ ID NO:6 VHLlopt: EVQLVESGGGLVQPGGSLRLSCAVSGGSISSGGYYWSWIRQAPGKGLEW VIGYIYYSGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC AR VD AR YD YVWGSYR YD AFDIWGQGTL VTVSS
  • MACPGFL W ALVISTCLEFSMASWAQSVLTQPPSVSAAPGQKVTISCSGSSS NIGNNYVSWYQQLPGTAPKLLIYDNNKRPSGIPDRFSGSKSGTSATLGITG LQTGDEADYYCGTWDSSLSAGWVFGGGTKLTVLGQPKAAPSVTLFPPS SEELQANKATLVCLISDFYPGAVTVAWKADSSPVKAGVETTTPSKQSNN KYAASSYLSLTPEQWKSHKSYSCQVTHEGSTVEKTVAPTECS
  • DNA sequence of CM-I heavy chain variable region agg tec ctg aga etc tec tgt gca gcc tct gga ttc ace ttc agt age tat ggc atg cac tgg gtc cgc cag get cca ggc aag ggg ctg gag tgg gtg gca gtt ata tea tat gat gga agt aat aaa tac tat gca gac tec gtg aag ggc cga ttc ace ate tec aga gac aat tec aag aac acg ctg tat ctg caa atg aac age ctg aga get gag gac acg get gtg tat tac tgt gcg aaa gac egg
  • DNA sequence of CM-I light chain variable region tec tat gtg ctg act cag cca ccc teg gtg tea gtg tec cca gga cag acg gcc agg ate ace tgc tct gga gat gca ttg cca aag caa tat get tat tgg tac cag cag aag cca ggc cag gcc ct gtg ctg gtg ata tat aaa gac agt gag agg cc tea ggg ate cct gag cga ttc tct ggc tec age tea ggg aca aca gtc acg ttg ace ate agt gga gtc cag gca gaa gac gag get gac tat tac
  • the DNA-PLOT sequence alignment program (available at http://www.dnaplot.org) identified the most homologous germ-line genes and to detect somatic mutations.
  • the CM-I heavy chain variable region (V H ) sequence is homologous to the IGHV3-3O/3-3O.5*Ol germ-line gene (IMGT No. X92214; Medline No.88283641; Berman et al., EMBO J.7:727-738, 1988) and the I H region of the IGHJ5*01 germ-line gene.
  • the CM-I light chain variable region (V L ) sequence is homologous to the IGLV3-25*03 germ-line gene (IMGT No. L29165; Medline No.94216813; Fang et al., J. Exp. Med.179:1445-1456, 1994) and the IL region of the IGL3*01 germ-line gene.
  • SAM-6 heavy chain variable region CDR3 ARDRLA V AGRPFDY.
  • SAM-6 light chain variable region CDRl, Ser, Asp, Lys, Leu, GIy, Asp Lys, CDR2, GIn, Asp, Ser, Lys, Arg, Pro, Ser, CDR3, gin, Ala, Trp, Asp, Ser, Ser, He, VaI, VaI.
  • Additional antibodies and fragments include SAM-6 variant heavy and light chain sequences.
  • Amino Acid SEQ ID NO: 13
  • nucleotide sequences of the variable region of the heavy chain (VH) of antibody SAM-6 cag gtg cag ctg gtg gag tct ggg gga ggc gtg gtc cag cct ggg 45
  • CDR3 acg get gtg tat tac tgt gcg aga gat egg tta gca gtg get ggt 315
  • IBTAl . .4 (F, B) QVQLVESGGG 1 WQPGRSLRL SRAASGFTFS SYAMHWVRQA PGKGLEWVAV ISYDGSNKYY ADSVKGRFTI (70)
  • Trp typically WYQ or WLQ or WFQ or WYL
  • SAM-6 1.6 VL (Used in all "A” scFv constructs, e.g., LlA scFv has this light chain sequence.) (SEQ ID NO:38)
  • SAM-6 2.6 VL (Used in all "B” scFv constructs, e.g., LIB scFv has this light chain sequence.) (SEQ ID NO:39)
  • PM-I heavy chain variable region CDRs 1, 2 and 3 are at amino acids 26-31, 49-51, and 88-95 of (SEQ ID NO:42)
  • age tea ggg aca atg gcc ace ttg act ate agt ggg gcc cag gtg gag 240 Ser Ser GIy Thr Met Ala Thr Leu Thr lie Ser GIy Ala Gin VaI GIu 65 70 75 80
  • PM-I light chain variable region CDRs 1, 2 and 3 are at amino acids 11-18, 36-43, and 82-90 of (SEQ ID NO.43).
  • gtc tea get att agt ggt agt ggt ggt agt aca tac tac gca gac tec 144 VaI Ser Ala lie Ser GIy Ser GIy GIy Ser Thr Tyr Tyr Ala Asp Ser
  • VaI Lys GIy Arg Phe Thr lie Ser Arg Asp Asn Ser Lys Asn Thr Leu 50 55 60 tat ctg caa atg aac age ctg aga gcc gag gac acg gcc gta tat tac 240 Tyr Leu GIn Met Asn Ser Leu Arg Ala GIu Asp Thr Ala VaI Tyr Tyr 65 70 75 80
  • PM- 2 heavy chain variable region CDRs 1, 2 and 3 are at amino acids 26-34, 52-58, and 97-103 of SEQ ID NO:44.
  • PM-2 light chain variable region CDRs 1, 2 and 3 are at amino acids 11-18, 36-43, and 82-100 of SEQ ID NO:45.
  • CM-2 heavy chain variable region CDRsI, 2 and 3 are at amino acids 26-34, 51-54, and 91-99 of SEQ ID NO:46.
  • cag tct gcc ctg act cag cct gcc tec gtg tct ggg tct cct gga cag 48 GIn Ser Ala Leu Thr GIn Pro Ala Ser VaI Ser GIy Ser Pro GIy GIn 1 5 10 15 teg ate ace ate tec tgc act gga ace age agt gac gtt ggt ggt tat 96 Ser lie Thr lie Ser Cys Thr GIy Thr Ser Ser Asp VaI GIy GIy Tyr 20 25 30
  • gag gac gag get gat tac tac tgc age tea aaa aga age age 288 GIn Ala GIu Asp GIu Ala Asp Tyr Tyr Cys Ser Ser Lys Arg Ser Ser 85 90 95
  • CM-2 light chain variable region CDRs 1, 2 and 3 are at amino acids 16-22, 40-47, and 86-100 of SEQ ID NO:47.
  • Nucleotide sequence of Norm-1 heavy chain variable region gaggtgcagc tgttggagtc tgggggaggc ttggtacagc ctggggggtc cctgagactc 60 tcctgtgcag cctctggatt cacctttagc agctatgcca tgagctgggt ccgccaggct 120 ccagggaagg ggctggagtg ggtctcagct attagtggta gtggtggtag cacatactac 180 gcagactccg tgaagggccg gttcaccatc tccagagaca attccaagaa cacgctgtat 240 ctgcaaatga acagcctgag agccgaggac acggccgtat atttccaagaa
  • CDRl of NORM-I heavy chain variable region spans nucleotides 91-105 which encode amino acids 31-35
  • CDR2 spans nucleotides 148-198 which encode amino acids 50-66
  • CDR 3 spans nucleotides 295-321 which encode amino acids 99-107.
  • the D-region spans nucleotides 297-319 and the J-region spans nucleotides 327-357.
  • Thr Ala Arg lie Thr Cys Ser GIy Asp Ala Leu Pro Lys Lys Tyr Ala
  • CDR 1 of the NORM- 1 light chain variable region spans nucleotides 67-99 which encode amino acids 23-33
  • CDR2 spans nucleotides 145- 165 which encode amino acids 49-55
  • CDR3 spans nucleotides 262-297 which encode amino acids 88-99.
  • the J-region spans nucleotides 291- 300.
  • Amino acid sequence of Norm-2 heavy chain variable region (SEQ ID NO:50): GIu VaI GIn Leu VaI GIu Ser GIy GIy GIy Leu VaI Lys Pro GIy GIy
  • nucleotide sequence of Norm-2 heavy chain variable region gaggtgcagc tggtggagtc tgggggaggc ctggtcaagc ctggggggtc cctgagactc 60 tcctgtgcag cctctggatt caccttcagt agctatagca tgaactgggt ccgccaggct 120 ccagggaagg ggctggagtg ggtctcatcc attagtagta gtagtagtta catatactac 180 gcagactcag tgaagggccg attcaccatc tccagagaca acgccaagaa ctcactgtat 240 ctgcaaatga acagcctgag agccgaggac acggctgt attactgtgc
  • CDRl of the NORM-2 heavy chain variable region spans nucleotides 91-105 which encode amino acids 31-35
  • CDR2 spans nucleotides 148-198 which encode amino acids 50-66
  • CDR3 spans nucleotides 295-324 which encode amino acids 99-108.
  • D-region spans nucleotides 297-300 and the J-region spans nucleotides 301-324.
  • Amino acid sequence of Norm-2 light chain variable region (SEQ ID NO:51): GIn Ser VaI Lou Thr GIn Pro Pro Ser VaI Ser GIy Ala Pro GIy GIn 1 5 10 15
  • Nucleotide sequence of Norm-2 light chain variable region cagtctgtgt tgacgcagcc gccctcagtg tctggggccc cagggcagag ggtcaccatc 60 tcctgcactg ggagcagctc caacatcggg gcaggttatg atgtacactg gtaccagcag 120 cttccaggaa cagcccccaa actcctcatc tatggtaaca gcaatcggcc ctcaggggtc 180 cctgaccggt tctctggctc caagtctggc acctcagcct ctggccat cactgggctc 240 caggctgagg atgaggctga ttattactgc cagtcctatg acagcagcc 240 caggc
  • CDRl of the NORM-2 light chain variable region spans nucleotides 67-108 which encode amino acids 23-36
  • CDR2 spans nucleotides 154-174 which encode amino acids 52-58
  • CDR3 spans nucleotides 271-303, which encode amino acids 91-101.
  • the J-region spans nucleotides 299-306.
  • Amino acid sequence of amplified B ARB 3 heavy chain variable region (SEQ ID NO:52):
  • BARB3 heavy chain CDR3 (by IMGT): gcacggacccgtcagccgtataaccagaatatggacgtc A R T R Q P Y N Q N M D V
  • BARB3-LC Light chain sequence SEQ ID NO:53, top
  • Z73650 IGLV8-61*01, bottom most homologous germeline gene
  • BARB3 light chain CDR3 (by IMGT): atg ttg tat gtg ggt agt gcc att tgg gtg M L Y V G S A I W V
  • Predicted CDRs of which there are three in each of heavy and light chain, are conveniently denoted herein as LC-CDRl, LC-CDR2 and LC- CDR3; and HC-CDRl, HC-CDR2 and HC-CDR3.
  • CDR sequences of each were determined by alignments with homologous germline sequences.
  • IMGT International ImMunoGeneTics Information System
  • MRC VBASE, MRC Centre for Protein Engineering sequence databases were used.
  • the IMGT database gives shorter CDR regions for lambda light chain.
  • BARB3 Heavy Chain CDRl GFSLSNAGMG; CDR2, IFSNDEK; and CDR3, ARTRQP YNQNMD V.
  • BARB3 Light Chain CDRl SGSVSPSYY; CDR2, STN; and CDR3, MLYVGSAIWV.
  • BARB3 Light Chain CDRl ALSSGSVSPSYYPS; CDR2, STNIRSS; and CDR3, MLYVGSAIW.
  • Amino acid sequence of amplified BARB4 heavy chain (SEQ ID NO:54):
  • Predicted CDRs of which there are three in each of heavy and light chain are conveniently denoted herein as LC-CDRl, LC-CDR2 and LC- CDR3; and HC-CDRl, HC-CDR2 and HC-CDR3.
  • CDR sequences of each of any of SEQ TD NOs: 1 to 58 can determined by alignments with homologous germline sequences (MRC VBASE, MRC Centre for Protein Engineering), or using Kabat numbering.
  • BARB4 Heavy Chain CDRl GFRFTTHGMH; CDR2, VISYNGRNKYYA; and CDR3, VRGDGYGD YGYF.
  • BARB4 Light Chain CDRl TGTYNYVS; CDR2, DVSRRSS; and CDR3, CSYGGTYLY.
  • Additional antibodies and fragments include BARB4 variant heavy and light chain sequences.
  • Amino acid sequence of BARB4 heavy chain (4.1 VH; SEQ ID NO:56):
  • binding when used in reference to an antibody or functional fragment, means that the antibody or functional fragment interacts at the molecular level with a corresponding epitope (antigenic determinant) present on a cell or an antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated Grp78, apoBlOO, LDL (e.g., oxLDL), VLDL, or deglycosylated LDL, Barb4 target, TAF-15 polypeptide), or compete for binding to a cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm- 1 target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosyl
  • Epitopes of antigens that comprise amino acids typically include relatively short sequences, e.g. about five to 15 amino acids in length. Epitopes can be contiguous or non-contiguous. A non-contiguous amino acid sequence epitope forms due to protein folding. Techniques for identifying epitopes are known to the skilled artisan and include screening overlapping oligopeptides for binding to antibody (for example, U.S. Patent No. 4,708,871 ), phage display peptide library kits, which are commercially available for epitope mapping (New England BioLabs).
  • Epitopes may also be identified by inference when epitope length peptide sequences are used to immunize animals from which antibodies that bind to the peptide sequence are obtained and can be predicted using computer programs, such as BEPITOPE (Odorico et al., /. MoI. Recognit. 16:20 (2003)).
  • the antibodies and functional fragments can inhibit, decrease or reduce cell growth or proliferation, or stimulate or induce cell death, lysis or apoptosis.
  • binding of an antibody, as represented by antibody produced by a hybridoma cell line set forth herein, or heavy and light chain variable region sequences set forth as in any of SEQ ID NOs: 1 to 58 to a neoplastic, tumor or cancer, or metastasis cell inhibits, decreases or reduces cell growth or proliferation, or stimulates or induces cell death, lysis or apoptosis.
  • binding of an antibody, as represented by any of SEQ ID NOs: 1 to 58, to cells inhibits, decreases or reduces cell growth or proliferation, or stimulates or induces cell death, lysis or apoptosis.
  • Antibodies and functional fragments can be structurally and/or functionally related to LM-I, CM-I, SAM-6, PM-I, PM-2, CM-2, Norm-1, Norm- 2, BARB3 or BARB4 as represented by a hybridoma cell line set forth herein; or a heavy and light chain variable region sequences in SEQ ID NOs: 1 to 58, in which include the heavy or light chain variable region sequence exhibits a degree of identity (less than 100%) to any of SEQ ID NOs: 1 to 58, or exhibits identity to a sequence within any of SEQ ID NOs: 1 to 58 (e.g., one or more CDRs).
  • Antibodies and functional fragments therefore include a heavy or a light chain variable region sequence with about 60% or more identity to a heavy or light chain sequence variable region of an antibody, as represented by a hybridoma, or heavy and light chain variable region sequences in any of SEQ ID NOs: 1 to 58, or a sequence within any of SEQ ID NOs: 1 to 58 (e.g., one or more CDRs).
  • antibodies or functional fragments include a heavy or a light chain with at least 65%, 70%, 75%, 80%, 85%, 90%, 95%, or more identity to a heavy chain variable region sequence of an antibody, , as represented by a hybridoma, or heavy and light chain variable region sequences in any of SEQ ID NOs: 1 to 58, or a sequence within any of SEQ ID NOs: 1 to 58 (e.g., one or more CDRs).
  • antibodies or functional fragments include a heavy or a light chain variable region sequence with at least 80-85%, 85- 90%, 90-95%, 95-100% identity to one or more CDRs of an antibody, as represented by a hybridoma, or a heavy and light chain variable region sequence set forth in any of SEQ ID NOs: 1 to 58.
  • an antibody or a functional fragment thereof includes a heavy or a light chain variable region sequence with 80%, 90%, or more, e.g., 95-100% identity to one, two or three CDRs in each heavy or light chain variable region sequences of an antibody, as represented by a hybridoma, or heavy and light chain variable region sequence in any of SEQ DD NOs: 1 to 58.
  • Antibodies and functional fragments of the invention therefore include those with at least partial sequence identity to a reference antibody, as represented by antibody produced by a hybridoma, or heavy and light chain variable region sequences set forth in any of SEQ ID NOs: 1 to 58.
  • the percent identity of such antibodies and functional fragments can be as little as 60%, or can be more (e.g., 65%, 70%, 75%, 80%, 85%, 90%, 95%, etc.).
  • the percent identity can extend over the entire sequence length, or a contiguous region or area within an antibody.
  • the length of the sequence sharing the percent identity is 5 or more contiguous amino acids, e.g., 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, etc. contiguous amino acids.
  • the length of the sequence sharing the percent identity is 25 or more contiguous amino acids, e.g., 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, etc. contiguous amino acids.
  • the length of the sequence sharing the percent identity is 35 or more contiguous amino acids, e.g., 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 45, 47, 48, 49, 50, etc., contiguous amino acids.
  • the length of the sequence sharing the percent identity is 50 or more contiguous amino acids, e.g., 50-55, 55-60, 60-65, 65-70, 70-75, 75-80, 80-85, 85-90, 90-95, 95-100, 100-110, etc.
  • the length of the sequence sharing the percent identity is equal to the length of any CDR of a variable region sequence, or a region outside the CDRs but within the variable region of a heavy or light chain sequence, such as an antibody represented by a hybridoma, or heavy and light chain variable region sequences set forth in any of SEQ ID NOs: 1 to 58.
  • identity and grammatical variations thereof, mean that two or more referenced entities are the same. Thus, where two antibody sequences are identical, they have the same amino acid sequence, at least within the referenced region or portion. Where two nucleic acid sequences are identical, they have the same polynucleotide sequence, at least within the referenced region or portion. The identity can be over a defined area (region or domain) of the sequence. An "area of identity” refers to a portion of two or more referenced entities that are the same. Thus, where two protein or nucleic acid sequences are identical over one or more sequence regions they share identity within that region. Exemplary identity are antibodies and functional fragments with an amino acid sequence with 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or more sequence identity to a reference antibody or functional fragment.
  • homology means that two or more referenced entities share at least partial identity over a given region or portion.
  • Areas, regions or domains of homology or identity mean that a portion of two or more referenced entities share homology or are the same. Thus, where two antibody sequences are identical over one or more sequence regions they share identity in these regions.
  • Substantial homology means that a molecule is structurally or functionally conserved such that it has or is predicted to have at least partial structure or function of one or more of the structures or functions (e.g., a biological function) of the reference molecule, or relevant/corresponding region or portion of the reference molecule to which it shares homology.
  • an antibody or functional fragment with substantial homology has or is predicted to have at least partial activity or function as the reference antibody.
  • an antibody as represented by an antibody produced by a hybridoma, or heavy and light chain variable region sequences set forth in any of SEQ ID NOs: 1 to 58, with one or more modifications (e.g., substitutions, deletions or additions) retain the ability to at least partially compete for binding of unmodified antibody, as represented by an antibody produced by a hybridoma, or heavy and light chain variable region sequences set forth in any of SEQ ID NOs: 1 to 58, to a cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated Grp78, apoBlOO, LDL (e.g., oxLDL),
  • apoBlOO, LDL e.g., oxLDL
  • VLDL e.g., adenosine
  • deglycosylated LDL e.g., Barb4 target, TAF- 15 polypeptide
  • the extent of identity (homology) between two sequences can be ascertained using a computer program and mathematical algorithm known in the ait.
  • Such algorithms that calculate percent sequence identity (homology) generally account for sequence gaps and mismatches over the comparison region or area.
  • a BLAST e.g., BLAST 2.0
  • search algorithm see, e.g., Altschul et al., /. MoI. Biol. 215:403 (1990), publicly available through NCBI
  • a BLASTP algorithm is typically used in combination with a scoring matrix, such as PAMlOO, PAM 250, BLOSUM 62 or BLOSUM 50.
  • FASTA e.g., FASTA2 and FASTA3
  • SSEARCH sequence comparison programs are also used to quantitate the extent of identity (Pearson et al., Proc. Natl. Acad. ScL USA 85:2444 (1988); Pearson, Methods MoI Biol. 132:185 (2000); and Smith et al., J. MoI. Biol. 147: 195 (1981)).
  • Programs for quantitating protein structural similarity using Delaunay-based topological mapping have also been developed (Bostick et al., Biochem Biophys Res Cotnmun. 304:320 (2003)).
  • Antibodies and functional fragments include those that retain at least one or more partial activities or functions of reference antibody.
  • the antigens to which the various antibodies set forth herein bind (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm- 2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated Grp78, apoBlOO, LDL (e.g., oxLDL), VLDL, or deglycosylated LDL, Barb4 target, TAF- 15 polypeptide), or compete for binding to a cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated
  • an antibody or functional fragment binds to one or more cells, to which an antibody set forth herein binds.
  • Such antibodies and functional fragments can have greater or less relative binding affinity for a cell or an antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated Grp78, apoBlOO, LDL (e.g., oxLDL), VLDL, or deglycosylated LDL, Barb4 target, TAF-15 polypeptide), or compete for binding to a cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6 target, Grp78, deg
  • Antibodies and functional fragments therefore include those that have greater than, about the same or less than the binding affinity of an antibody set forth herein, as represented by antibody produced by a hybridoma, or heavy and light chain variable region sequences set forth in any of SEQ ID NOs: 1 to 58, for binding to a cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated Grp78, apoBlOO, LDL (e.g., oxLDL), VLDL, or deglycosylated LDL, Barb4 target, TAF-15 polypeptide), or compete for binding to a cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, L
  • an antibody or functional fragment may have an affinity greater or less than 2-5, 5-10, 10-100, 100-1000 or 1000-10,000-fold affinity, or any numerical value or range within or encompassing such values, than an antibody set forth herein for binding to a cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated Grp78, apoBlOO, LDL (e.g., oxLDL), VLDL, or deglycosylated LDL, Barb4 target, TAF-15 polypeptide), or compete for binding to a cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6
  • Such antibodies can compete for binding of a reference antibody (e.g., LM-I, CM-I, CM-2, PM-I, PM-2, Norm-1, Norm-2, SAM-6, Barb3, Barb4, etc.) to a cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated Grp78, apoBlOO, LDL (e.g., oxLDL), VLDL, or deglycosylated LDL, Barb4 target, TAF- 15 polypeptide), or compete for binding to a cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein,
  • Binding affinity can be determined by association (K a ) and dissociation (IQ) rate.
  • Equilibrium affinity constant, K is the ratio of K a /IQ.
  • Association (K a ) and dissociation (K d ) rates can be measured using surface plasmon resonance (SPR) (Rich and Myszka, Curr. Opin. Biotechnol. 11:54 (2000); Englebienne, Analyst. 123:1599 (1998)). Instrumentation and methods for real time detection and monitoring of binding rates are known and are commercially available (BiaCore 2000, Biacore AB, Upsala, Sweden; and Malmqvist, Biochem. Soc. Trans. 27:335 (1999)).
  • Additional specific non-limiting antibodies and functional fragments have binding affinity for a cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm- 1 target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated Grp78, apoBlOO, LDL (e.g., oxLDL), VLDL, or deglycosylated LDL, Barb4 target, TAF- 15 polypeptide), or compete for binding to a cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated Grp78, apoBlOO, LDL (e.g., oxLDL), VLD
  • binding affinity for is less than 5xlO "2 M, 10 "2 M, 5xlO "3 M, 10 "3 M 5xlO "4 M, 10 "4 M 5x10 5 M, 10 5 M 5xlO "6 M, 10 6 M 5xlO 7 M, 10 "7 M 5x10 8 M, 10 8 M 5xlO ⁇ 9 M, 10 "9 M SxIO- 10 M, 10 "10 M 5x10 " “ M, 10 " “ M 5xl0 “l2 M, 10 "12 M 5xlO "13 M. 10 "13 M 5xlO "14 M, 10 ⁇ 14 M 5xlCT 15 M, and ICT 15 M.
  • an antibody or functional fragment has a binding affinity within about K d 10 ⁇ 5 M to about Kd 10 13 M for binding to a neoplastic, cancer, tumor or metastatic cell.
  • Antibodies and functional fragments that bind to a cell or antigen to which an antibody set forth herein binds, or that competes with an antibody set forth herein for binding to a cell or antigen can have greater or less relative cell proliferation inhibiting or reducing activity, or greater or less relative cell apoptosis inducing or stimulating activity than particular antibody set forth herein.
  • Antibodies and functional fragments therefore include those that bind to a cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated Grp78.
  • apoBlOO LDL (e.g., oxLDL), VLDL, or deglycosylated LDL, Barb4 target, TAF- 15 polypeptide), or compete for binding to a cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated Grp78, apoBlOO, LDL (e.g., oxLDL), VLDL, or deglycosylated LDL, Barb4 target, TAF- 15 polypeptide), and compete from binding of a reference antibody (e.g., (e.g., LM-I, CM-I, CM-2, PM-I, PM- 2, Norm-1, Norm-2, SAM-6, Barb3, Barb4, etc.) as well as antibodies that have greater or less relative cell proliferation inhibiting or
  • Antibodies and functional fragments therefore include those that have a sequence distinct from antibody set forth herein, but that retain one or more activities or functions, at least in part, of the reference antibody set forth herein. Modified antibodies and functional fragments retain, at least a part of an activity or function of unmodified or reference antibody, or functional fragment.
  • such an antibody or a functional fragment thereof retains at least partial activity or function of intact full length antibody, such as competes for binding to a cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated Grp78, apoBlOO, LDL (e.g., oxLDL), VLDL, or deglycosylated LDL, Barb4 target, TAF- 15 polypeptide), to which the antibody binds; inhibits or reduces proliferation of a cell in which the antibody inhibits or reduces proliferation; stimulates or induces death, lysis or apoptosis of a cell; cell growth or proliferation is inhibited, decreased or reduced at least 20%, 30%, 40%, 50%, 60%, 75%, or more relative to a control (untreated) cell; or cell
  • modified proteins and grammatical variations thereof, means that the composition deviates from a reference composition.
  • modified proteins, nucleic acids and other compositions may have greater or less activity than or a distinct function from a reference unmodified protein, nucleic acid, or composition.
  • Modifications which include substitutions, additions and deletions, can also be referred to as "variants.”
  • Specific non-limiting examples of amino acid variants include antibody fragments and subsequences.
  • Exemplary antibody subsequences and fragments include a portion of the antibody, that at least partially competes with intact antibody for binding to a cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated Grp78, apoB lOO, LDL (e.g., oxLDL), VLDL, or deglycosylated LDL, Barb4 target, TAF- 15 polypeptide), or that retains at least partial binding activity to a cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2
  • fragment means a portion of the full length molecule.
  • a fragment or subsequence of an antibody has one or more less amino acids than a full length intact reference antibody (e.g. one or more internal or terminal amino acid deletions from either amino or carboxy-termini of heavy or light chain variable or constant regions).
  • a nucleic acid fragment has at least one less nucleotide than a full length comparison nucleic acid sequence. Fragments therefore can be any length up to the full length native molecule.
  • a functional fragment when referring to an antibody refers to a portion of an antibody with a function or activity.
  • a functional fragment can retain one or more partial functions or activities as an intact reference antibody, e.g., a function or activity of an antibody produced by hybridoma or comprising heavy and light chain sequences set forth in any of SEQ ID NOs: 1 to 58.
  • Antibody fragments can include all or a portion of heavy or light chain variable region(s) (e.g., one or more CDRs, such as CDRl, CDR2 or CDR3) alone or in combination with all or a portion of one or more of the following: hinge region, CHl, CH2, and CH3 domains. Also included are antigen-binding subsequences of any combination of of heavy or light chain variable region(s) (e.g., one or more CDRs, such as CDRl, CDR2 or CDR3) with a hinge region, CHl, CH2, and CH3 domains
  • Exemplaiy antibody subsequences and fragments of the invention include Fab, Fab', F(ab'> 2 , Fv, Fd, single-chain Fv (scFv), disulfide- linked Fvs (sdFv), V L and V H domain fragments, trispecific (Fab 3 ), bispecific (Fab 2 ), diabody ((V L -V H ) 2 or (V H -V 1 ) 2 ), triabody (trivalent), tetrabody (tetravalent), minibody ((SCF V -C H 3> 2 ), bispecific single-chain Fv (Bis-scFv), IgGdeltaCH2, scFv-Fc and (scFv) 2 -Fc.
  • Such subsequences and fragments can have binding affinity as the full length antibody, the binding specificity as the full length antibody, oi one or moie activities or functions of as a full length antibody.
  • Antibody subsequences and fragments can be combined.
  • a VL or VH subsequences can be joined by a linker sequence thereby forming a V L -V H chimera.
  • a heavy chain variable sequence of an antibody can be combined with a light chain variable sequence of an antibody.
  • a combination of single-chain Fvs (scFv) subsequences can be joined by a linker sequence thereby forming a scFv - scFv chimera.
  • Antibody subsequences and fragments include single-chain antibodies or variable region(s) alone or in combination with all or a portion of other subsequences.
  • Modified proteins further include ammo acid substitutions.
  • Substitutions can be conservative or non-conservative and may be in a constant or variable (e.g., hypervariable, such as CDR or FR) region of an antibody.
  • a modified antibody as represented by an antibody produced by a hybridoma, or heavy and light chain sequences set forth in any of SEQ ID NOs: 1 to 58, has one or a few conservative or non-conservative amino acid substitutions.
  • Antibody structural determinants that contribute to antigen binding such as complemetarity determining regions (CDR, of which there are three in each heavy and light chain sequence, conveniently denoted as HC-CDRl, HC-CDR2 and HC-CDR3; and LC-CDRl, LC-CDR2 and LC-CDR3) within hypervariable regions are known to the skilled artisan.
  • CDR complemetarity determining regions
  • Antibodies and subsequences thereof include those in which one or more CDR sequences have sufficient sequence identity to a reference antibody, so as to retain at least partial function or activity of reference antibody, e.g., cell or antigen binding, binding affinity (e.g., IQ), cell proliferation inhibition, or stimulating or inducing cell apoptosis, etc.
  • reference antibody e.g., cell or antigen binding, binding affinity (e.g., IQ), cell proliferation inhibition, or stimulating or inducing cell apoptosis, etc.
  • a "conservative substitution” is the replacement of one amino acid by a biologically, chemically or structurally similar residue.
  • Biologically similar means that the substitution does not destroy a biological activity, e.g., cell binding or cell proliferation inhibting or apoptosis inducing or stimulating activity.
  • Structurally similar means that the amino acids have side chains with similar length, such as alanine, glycine and serine, or a similar size.
  • Chemical similarity means that the residues have the same charge or are both hydrophilic or hydrophobic.
  • Particular examples include the substitution of one hydrophobic residue, such as isoleucine, valine, leucine or methionine for another, or the substitution of one polar residue for another, such as the substitution of arginine for lysine, glutamic for aspartic acids, or glutamine for asparagine, serine for threonine, and the like.
  • a heavy or light chain hypervariable region sequence or a region therein, such as a CDR (CDRl, CDR2 or CDR3) or FR will have 1-10, 1-5, 1-3 or fewer (e.g., 1 or 2) amino acid substitutions.
  • an amino acid substitution within a heavy or light chain hypervariable region sequence is not within more than one CDR.
  • a substitution within a heavy or light chain hypervariable region sequence is not within a CDR
  • a substitution within a hyperva ⁇ able region sequence is not within an FR
  • the effect of a given modification can be readily assayed in order to identify antibodies and functional fragments retaining at least a part of the cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated Grp78, apoB lOO, LDL (e.g , oxLDL), VLDL, or deglycosylated LDL, Barb4 target, TAF- 15 polypeptide), or compete for binding to a cell or antigen (e g , CM- 1 target, CM-2 target, Pm- 1 target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated Grp78, apoBlOO
  • an amino acid substitution in a variable region (e g , within or outside of CDRl, CDR2 or CDR3 of an antibody), as represented by an antibody produced by a hyb ⁇ doma, or heavy and light chain sequences set forth in any of SEQ ID NOs 1 to 58 can be assayed for cell or antigen binding, cell proliferation inhbitmg or reducing activity, inducing or stimulating cell death, lysis or apoptosis, etc.
  • Predictive models based upon OSAR can in turn be used to predict the effect of substitutions (mutations)
  • the effect of mutations on the association and dissociation rate of an antibody interacting with its antigen has been used to construct quantitative predictive models for both kinetic (K d and IQ) constants, which m turn is used to predict the effect of other mutations on the antibody (De Genst et al , J BwI Chem 277:29897 (2002)).
  • K d and IQ kinetic constants
  • Amino acid substitutions may be with the same amino acid, except that a naturally occurring L-amino acid is substituted with a D-form amino acid.
  • Modifications therefore include one or more D-amino acids substituted for L-amino acids, or mixtures of D-amino acids substituted for L-amino acids.
  • Modifications also include structural and functional analogues, for example, peptidomimetics having synthetic or non-natural amino acids or amino acid analogues and derivatized forms.
  • Modified forms further include derivatized sequences, for example, amino acids in which free amino groups form amine hydrochlorides, p- toluene sulfonyl groups, carbobenzoxy groups; the free carboxy groups from salts, methyl and ethyl esters; free hydroxl groups that form O-acyl or O-alkyl derivatives, as well as naturally occurring amino acid derivatives, for example, 4- hydroxyproline, for proline. 5-hydroxylysine for lysine, homoserine for serine, ornithine for lysine, etc. Modifications can be produced using methods known in the art ⁇ e.g., PCR based site-directed, deletion and insertion mutagenesis, chemical modification and mutagenesis, cross-linking, etc.).
  • Modified forms include additions and insertions.
  • an addition can be the covalent or non-covalent attachment of any type of molecule to a protein ⁇ e.g., antibody), nucleic acid or other composition.
  • additions and insertions confer a distinct function or activity.
  • Additions and insertions include fusion (chimeric) polypeptide or nucleic acid sequences, which is a sequence having one or more molecules not normally present in a reference native (wild type) sequence covalently attached to the sequence.
  • a particular example is an amino acid sequence of another protein ⁇ e.g., antibody) to produce a multifunctional protein ⁇ e.g., multispecific antibody).
  • a heterologous domain can consist of any of a variety of different types of small or large functional moieties. Such moieties include nucleic acid, peptide, carbohydrate, lipid or small organic compounds, such as a drug ⁇ e.g., a cell proliferative agent), metals (gold, silver), etc.
  • a heterologous domains can be an amino acid addition or insertion.
  • Particular non-limiting examples of heterologous domains include, for example, tags, detectable labels and cytotoxic agents.
  • tags and detectable labels include enzymes (horseradish peroxidase, urease, catalase, alkaline phosphatase, beta-galaclosidase, chloramphenicol transferase); enzyme substrates; ligands (e.g., biotin); receptors (avidin); radionuclides (e.g., C 14 , S 35 , P 32 , P 33 , H 3 , 1 125 , 1 131 , gallium-67 and 68, scantium- 47, indium- 111, radium-223); T7-, His-, myc-, HA- and FLAG-tags; electron- dense reagents; energy transfer molecules; paramagnetic labels; fluorophores (fluorescein, rhodamine, phycoerthrin); chromophores; chemi-luminescent (imidazole, luciferase); and bio-luminescent agents.
  • enzymes e.g
  • heterologous domains include, for example, anti-cell proliferative agents (e.g., anti-neoplastic, anti-tumor or anticancer, or anti-metastasis agents). Specific non-limiting examples of anti-cell proliferative agents are disclosed herein and known in the art.
  • Linker sequences may be inserted between the protein (e.g., antibody), nucleic acid, or other composition and the addition or insertion (e.g., heterologous domain) so that the two entities maintain, at least in part, a distinct function or activity.
  • Linker sequences may have one or more properties that include a flexible structure, an inability to form an ordered secondary structure or a hydrophobic or charged character which could promote or interact with either domain.
  • Amino acids typically found in flexible protein regions include GIy, Asn and Ser. Other near neutral amino acids, such as Thr and Ala, may also be used in the linker sequence.
  • the length of the linker sequence may vary (see, e.g., U.S. Patent No. 6,087,329).
  • Linkers further include chemical cross-linking and conjugating agents, such as sulfo-succinimidyl derivatives (sulfo-SMCC, sulfo- SMPB), disuccinimidyl suberate (DSS), disuccinimidyl glutarate (DSG) and disuccinimidyl tartrate (DST).
  • chemical cross-linking and conjugating agents such as sulfo-succinimidyl derivatives (sulfo-SMCC, sulfo- SMPB), disuccinimidyl suberate (DSS), disuccinimidyl glutarate (DSG) and disuccinimidyl tartrate (DST).
  • compositions so separated are substantially free of one or more materials with which they normally associate with in nature, for example, one or more protein, nucleic acid, lipid, carbohydrate, cell membrane.
  • an isolated composition is substantially separated from other biological components in the cell of the organism in which the composition naturally occurs, or from the artificial medium in which it is produced (e.g., synthetically or through cell culture).
  • an isolated polypeptide is substantially separated from other polypeptides and nucleic acid and does not include a library of polypeptides or polynucleotides present among millions of polypeptide or nucleic acid sequences, such as a polypeptide, genomic or cDNA library, for example.
  • An isolated nucleic acid is substantially separated from other polypeptides and nucleic acid and does not include a library of polypeptides or polynucleotides present among millions of polypeptide or nucleic acid sequences, such as a polypeptide, genomic or cDNA library, for example.
  • isolated does not exclude alternative physical forms of the composition, for example, an isolated protein could include protein multimers, post-translational modifications (e.g., glycosylation, phosphorylation) or derivatized forms.
  • purified used as a modifier of a composition refers to a composition free of most or all of the materials with which it typically associates with in nature.
  • a protein separated from cells is considered to be substantially purified when separated from cellular components by standard methods while a chemically synthesized nucleic acid sequence is considered to be substantially purified when separated from its chemical precursors. Purified therefore does not require absolute purity.
  • a “purified” composition can be combined with one or more other molecules. Thus, the term “purified” does not exclude combinations of compositions.
  • Proteins and nucleic acid include proteins and nucleic acids produced by standard purification methods. The term also includes proteins and nucleic acids produced by recombinant expression in a host cell as well as chemical synthesis. “Purified” can also refer to a composition in which the level of contaminants is below a level that is acceptable to a regulatory agency for administration to a human or non-human animal, for example, the Food and Drug administration (FDA).
  • FDA Food and Drug administration
  • Substantial purity can be at least about 60% or more of the molecule by mass. Purity can also be about 70% or 80% or more, and can be greater, for example, 90% or more. Purity can be less, for example, in a pharmaceutical carrier the amount of a molecule by weight % can be less than 60% but the relative proportion of the molecule compared to other components with which it is normally associated with will be greater. Purity can be determined by any appropriate method, including, for example, UV spectroscopy, chromatography (e.g., HPLC, gas phase), gel electrophoresis (e.g., silver or coomassie staining) and sequence analysis (peptide and nucleic acid).
  • chromatography e.g., HPLC, gas phase
  • gel electrophoresis e.g., silver or coomassie staining
  • sequence analysis peptide and nucleic acid
  • polyclonal and monoclonal antibodies are known in the art.
  • an antigen or an immunogenic fragment thereof optionally conjugated to a carrier such as keyhole limpet hemocyanin (KLH) or ovalbumin (e.g., BSA), or mixed with an adjuvant such as Freund's complete or incomplete adjuvant, and used to immunize an animal.
  • KLH keyhole limpet hemocyanin
  • BSA ovalbumin
  • an adjuvant such as Freund's complete or incomplete adjuvant
  • splenocytes from immunized animals that respond to antigen can be isolated and fused with myeloma cells.
  • Monoclonal antibodies produced by the hybridomas can be screened for reactivity.
  • Proteins and antibodies, subsequences and fragments thereof, as well as other modified sequences can be produced by genetic methodology. Such techniques include expression of all or a part of the gene encoding the protein or antibody into a host cell such as Cos cells or E. coli. Such host cells can express full length or a fragment, for example, an scFv (see, e.g., Whitlow et al, In: Methods: A Companion to Methods in Enzvmologv 2:97 (1991), Bird et ai, Science 242:423 (1988); and U.S. Patent No. 4,946,778).
  • scFv see, e.g., Whitlow et al, In: Methods: A Companion to Methods in Enzvmologv 2:97 (1991), Bird et ai, Science 242:423 (1988); and U.S. Patent No. 4,946,778).
  • Antibodies and functional fragments, and nucleic acid sequences can also be produced by chemical synthesis using methods known to the skilled artisan, for example, an automated peptide synthesis apparatus (see, e.g., Applied Biosystems, Foster City, CA).
  • Antibodies and functional fragments can be screened or selected using various assays know in the art, such as enzyme linked immunosorbent assay (ELISA), phage display, protein-mRNA link via ribosome and mRNA display, display on yeast, bacteria, mammalian cells or retroviruses, microbead via in vitro compartmentalization, protein-DNA display, growth selection via yeast 2-hybrid, protein fragment complementation (Hoogenboom, R., Nature Biotechnoi 23:1105 (2005)).
  • ELISA enzyme linked immunosorbent assay
  • Cells or antigen e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm-I target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated G ⁇ 78, apoB lOO, LDL (e.g., oxLDL), VLDL, or deglycosylated LDL, Barb4 target, TAF-15 polypeptide), or compete for binding to a cell or antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target, Barb3 target, Lm- 1 target, NONO/nmt55 protein, SAM-6 target, Grp78, deglycosylated Grp78, apoBlOO, LDL (e.g., oxLDL), VLDL, or deglycosylated LDL, Barb4 target
  • Animals that may be immunized include mice, rats, rabbits, goats, sheep, cows or steer, guinea pigs or primates.
  • Initial and any optional subsequent immunization may be through intravenous, intraperitoneal, intramuscular, or subcutaneous routes.
  • Subsequent immunizations may be at the same or at different concentrations of cell or antigen preparation, and may be at regular or irregular intervals.
  • Animals include those genetically modified to include human IgG gene loci, which can therefore be used to produce human antibodies.
  • Transgenic animals with one or more human immunoglobulin genes that do not express endogenous immunoglobulins are described, for example in, U.S. Patent No. 5,939,598. Additional methods for producing human polyclonal antibodies and human monoclonal antibodies are described (see, e.g., Kuroiwa et ah, Nat. Biotechnoi. 20:889 (2002); WO 98/24893; WO 92/01047; WO 96/34096; WO 96/33735; U.S. Patent Nos.
  • Antibodies can also be generated using other techniques including hybridoma, recombinant, and phage display technologies, or a combination thereof (see U.S. Patent Nos. 4,902,614, 4,543,439, and 4,41 1 ,993; see, also Monoclonal Antibodies, Hybridomas: A New Dimension in Biological Analyses, Plenum Press, Kennett, McKearn. and Bechtol (eds.), 1980, and Harlow et al, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, 2nd ed. 1988).
  • Antibody subsequences and fragments can be prepared by proteolytic hydrolysis of the antibody, for example, by pepsin or papain digestion of whole antibodies.
  • Antibody subsequences and fragments produced by enzymatic cleavage with pepsin provide a 5S fragment denoted F(ab') 2 . This fragment can be further cleaved using a thiol reducing agent to produce 3.5S Fab' monovalent fragments.
  • an enzymatic cleavage using pepsin produces two monovalent Fab' fragments and the Fc fragment directly (see, e.g., U.S. Patent Nos.
  • Single-chain Fvs and antibodies can be produced as described in U.S. Patent Nos. 4,946,778 and 5,258,498; Huston et al, Methods Enzymol. 203:46 (1991); Shu et al, Proc. Natl. Acad. ScL USA 90:7995 (1993); and Skerra et al. Science 240: 1038 (1988).
  • Other methods of cleaving antibodies such as separation of heavy chains to form monovalent light-heavy chain fragments, further cleavage of fragments, or other enzymatic or chemical may also be used.
  • Modified antibodies and functional fragments having altered characteristics can be produced using methods known to the skilled artisan art.
  • affinity maturation techniques can be used to improve antibody binding affinity (US 2004/0162413 Al; U.S. Patent Nos. 6,656,467, 6,531,580, 6,590,079 and 5,955,358; Fiedler et al., Protein Eng. 15:931 (2002); Pancook et al., Hybrid. Hybridomics 20:383 (2001); Daugherty et al., Protein Eng. 11:825 (1998); Wu et al., Proc. Nat'l Acad. ScL USA 95:6037 (1998); and Osbourn et al., Immunotechnology 2: 181 (1996)).
  • Antibodies can be humanized using a variety of techniques known in the art including, for example, CDR-grafting (EP 239,400; W091/09967; U.S. Patent Nos. 5,225,539; 5,530,101; and 5,585,089), veneering or resurfacing (EP 592,106; EP 519,596: Padlan, Molecular Immunol. 28:489 (1991); Studnicka et al, Protein Engineering 7:805 (1994); Roguska. et al, Proc. Nat'l. Acad. ScL USA 91:969 (1994)), and chain shuffling (U.S. Patent No. 5,565,332). Human consensus sequences (Padlan, MoI Immunol.
  • Suitable techniques that additionally may be employed in antibody methods include affinity purification, non-denaturing gel purification, HPLC or RP-HPLC, size exclusion, purification on protein A column, or any combination of these techniques.
  • the antibody isotype can be determined using an ELISA assay, for example, a human Ig can be identified using mouse Ig- absorbed anti-human Ig.
  • Proteins such as antibodies that include amino acid substitutions, additions or deletions can be encoded by a nucleic acid. Consequently, nucleic acid sequences encoding proteins that include amino acid substitutions, additions or deletions may be used to produce antibodies and functional fragments.
  • nucleic acid and “polynucleotide” and the like refer to at least two or more ribo- or deoxy-ribonucleic acid base pairs (nucleotides) that are linked through a phosphoester bond or equivalent.
  • Nucleic acids include polynucleotides and polynucleosides. Nucleic acids include single, double or triplex, circular or linear, molecules. Exemplary nucleic acids include but are not limited to: RNA, DNA, cDNA, genomic nucleic acid, naturally occurring and non naturally occurring nucleic acid, e.g., synthetic nucleic acid.
  • Nucleic acids can be of various lengths. Nucleic acid lengths typically range from about 20 nucleotides to 20 Kb, or any numerical value or range within or encompassing such lengths, 10 nucleotides to 10Kb, 1 to 5 Kb or less, 1000 to about 500 nucleotides or less in length. Nucleic acids can also be shorter, for example, 100 to about 500 nucleotides, or from about 12 to 25, 25 to 50, 50 to 100, 100 to 250, or about 250 to 500 nucleotides in length, or any numerical value or range or value within or encompassing such lengths.
  • a nucleic acid sequence has a length from about 10-20, 20-30, 30-50, 50-100, 100-150, 150-200, 200-250, 250-300, 300-400, 400-500, 500-1000, 1000-2000, nucleotides, or any numerical value or range within or encompassing such lengths.
  • Shorter polynucleotides are commonly referred to as “oligonucleotides” or “probes” of single- or double-stranded DNA. However, there is no upper limit to the length of such oligonucleotides.
  • Polynucleotides include L- or D-forms and mixtures thereof, which additionally may be modified to be resistant to degradation when administered to a subject. Particular examples include 5' and 3' linkages resistant to endonucleases and cxonucleases present in various tissues or fluids of a subject.
  • Nucleic acid sequences further include nucleotide and nucleoside substitutions, additions and deletions, as well as derivatized forms and fusion/chimeric sequences (e.g., encoding recombinant polypeptide).
  • nucleic acids include sequences and subsequences degenerate with respect to nucleic acids that encode the same polypeptide sequence, modified forms and variants thereof.
  • Other examples are nucleic acids complementary to a sequence that encodes a protein, antibody, or fragment thereof.
  • Nucleic acid deletions can have from about 10 to 25, 25 to 50 or 50 to 100 nucleotides. Such nucleic acids are useful for expressing polypeptide subsequences, for genetic manipulation (as primers and templates for PCR amplification), and as probes to detect the presence or an amount of a sequence encoding a protein (e.g., via hybridization), in a cell, culture medium, biological sample (e.g., tissue, organ, blood or serum), or in a subject.
  • a sequence encoding a protein e.g., via hybridization
  • Nucleic acids can be produced using various standard cloning and chemical synthesis techniques. Techniques include, but are not limited to nucleic acid amplification, e.g., polymerase chain reaction (PCR), with genomic DNA or cDNA targets using primers (e.g., a degenerate primer mixture) capable of annealing to antibody encoding sequence. Nucleic acids can also be produced by chemical synthesis (e.g., solid phase phosphoramidite synthesis) or transcription from a gene.
  • PCR polymerase chain reaction
  • primers e.g., a degenerate primer mixture
  • Nucleic acids can also be produced by chemical synthesis (e.g., solid phase phosphoramidite synthesis) or transcription from a gene.
  • sequences produced can then be translated in vitro, or cloned into a plasmid and propagated and then expressed in a cell (e.g., a host cell such as yeast or bacteria, a eukaryote such as an animal or mammalian cell or in a plant).
  • a cell e.g., a host cell such as yeast or bacteria, a eukaryote such as an animal or mammalian cell or in a plant.
  • a vector that comprises nucleic acid sequences of the invention.
  • a vector includes a nucleic acid sequence encoding an antibody or functional fragment as set forth herein, hi another embodiment, a vector includes a nucleic acid sequence encoding a recombinant protein comprising an antibody or functional fragment thereof.
  • Vectors include viral, prokaryotic (bacterial) and eukaryotic (plant, fungal, mammalian) vectors.
  • Vectors can be used for expression of nucleic acids in vitro or in vivo.
  • Such vectors referred to as "expression vectors,” are useful for introducing nucleic acids, including nucleic acids that encode antibody, and expressing the encoded protein or inhibitory nucleic acid (e.g., in solution or in solid phase), in cells or in a subject in vivo.
  • Vectors can also be used for manipulation of nucleic acids.
  • "cloning vectors” can be employed, and to transcribe or translate the inserted nucleic acid.
  • a vector generally contains an origin of replication for propagation in a cell in vitro or in vivo.
  • Control elements including expression control elements, present within a vector, can be included to facilitate transcription and translation, as appropriate.
  • Vectors can include a selection marker.
  • a "selection marker” is a gene that allows for the selection of cells containing the gene. "Positive selection” refers to a process in which cells that contain the selection marker survive upon exposure to the positive selection. Drug resistance is one example of a positive selection marker-cells containing the marker will survive in culture medium containing the selection drug, and cells lacking the marker will die. Selection markers include drug resistance genes such as neo, which confers resistance to G418; hygr, which confers resistance to hygromycin; and puro, which confers resistance to puromycin. Other positive selection marker genes include genes that allow identification or screening of cells containing the marker.
  • GFP and GFP-like chromophores genes for fluorescent proteins (GFP and GFP-like chromophores, luciferase), the lacZ gene, the alkaline phosphatase gene, and suiface markers such as CD8, among others.
  • Negative selection refers to a process in which cells containing a negative selection marker are killed upon exposure to an appropriate negative selection agent.
  • cells which contain the herpes simplex virus-thymidine kinase (HSV-tk) gene (Wigler et ai, Cell 11:223 (1977)) are sensitive to the drug gancyclovir (GANC).
  • GANC drug gancyclovir
  • the gpt gene renders cells sensitive to 6-thioxanthine.
  • Viral vectors include those based upon retroviral (lentivirus for infecting dividing as well as non-dividing cells), foamy viruses (U.S. Patent Nos. 5,624,820, 5,693,508, 5,665,577, 6,013,516 and 5,674,703; WO92/05266 and WO92/14829), adenovirus (U.S. Patent Nos. 5,700,470, 5,731 ,172 and 5,928,944), adeno-associated virus (AAV) (U.S. Patent No. 5,604,090), herpes simplex virus vectors (U.S. Patent No. 5,501,979), cytomegalovirus (CMV) based vectors (U.S. Patent No.
  • Adenovirus efficiently infects slowly replicating and/or terminally differentiated cells and can be used to target slowly replicating and/or terminally differentiated cells.
  • Additional viral vectors useful for expression include parvovirus, Norwalk vims, coronaviruses, paramyxo- and rhabdoviruses, togavirus (e.g., Sindbis virus and semliki forest virus) and vesicular stomatitis virus (VSV).
  • parvovirus Norwalk vims
  • coronaviruses coronaviruses
  • paramyxo- and rhabdoviruses e.g., Sindbis virus and semliki forest virus
  • VSV vesicular stomatitis virus
  • a nucleic acid can be expressed when the nucleic acid is operably linked to an expression control element.
  • operably linked refers to a physical or a functional relationship between the elements referred to that permit them to operate in their intended fashion.
  • an expression control element "operably linked" to a nucleic acid means that the control element modulates nucleic acid transcription and as appropriate, translation of the transcript.
  • expression control element refers to nucleic acid that influences expression of an operably linked nucleic acid. Promoters and enhancers are particular non-limiting examples of expression control elements.
  • a "promoter sequence” is a DNA regulatory region capable of initiating transcription of a downstream (3' direction) sequence. The promoter sequence includes nucleotides that facilitate transcription initiation. Enhancers also regulate gene expression, but can function at a distance from the transcription start site of the gene to which it is operably linked. Enhancers function at either 5' or 3' ends of the gene, as well as within the gene (e.g., in introns or coding sequences).
  • Additional expression control elements include leader sequences and fusion partner sequences, internal ribosome binding sites (IRES) elements for the creation of multigene, or polycistronic, messages, splicing signal for introns, maintenance of the correct reading frame of the gene to permit in-frame translation of mRNA, polyadenylation signal to provide proper polyadenylation of the transcript of interest, and stop codons.
  • IRS internal ribosome binding sites
  • Expression control elements include “constitutive" elements in which transcription of an operably linked nucleic acid occurs without the presence of a signal or stimuli.
  • Expression control elements that confer expression in response to a signal or stimuli which either increase or decrease expression of operably linked nucleic acid, are “regulatable.”
  • a regulatable element that increases expression of operably linked nucleic acid in response to a signal or stimuli is referred to as an “inducible element.”
  • a regulatable element that decreases expression of the operably linked nucleic acid in response to a signal or stimuli is referred to as a "repressible element" (i.e., the signal decreases expression; when the signal is removed or absent, expression is increased).
  • Expression control elements include elements active in a particular tissue or cell type, referred to as “tissue-specific expression control elements.” Tissue-specific expression control elements are typically more active in specific cell or tissue types because they are recognized by transcriptional activator proteins, or other transcription regulators active in the specific cell or tissue type, as compared to other cell or tissue types.
  • Tissue-specific expression control elements include promoters and enhancers active in hyperproliferative cells, such as cell proliferative disorders including neoplasias, tumors and cancers, and metastasis.
  • promoters are hexokinase II, COX-2, alpha- fetoprotein, carcLnoembryonic antigen, DE3/MUC1, prostate specific antigen, C- erB2/neu, telomerase reverse transcriptase and hypoxia-responsive promoter.
  • constitutive promoters include T7, as well as inducible promoters such as pL of bacteriophage ⁇ , plac, ptrp, ptac (ptrp-lac hybrid promoter).
  • constitutive or inducible promoters e.g., ecdysone
  • constitutive promoters include, for example, ADH or LEU2 and inducible promoters such as GAL (see, e.g., Ausubel et al., In: Current Protocols in Molecular Biology, Vol. 2, Ch. 13, ed., Greene Publish. Assoc.
  • constitutive promoters of viral or other origins may be used.
  • SV40, or viral long terminal repeats (LTRs) and the like, or inducible promoters derived from the genome of mammalian cells ⁇ e.g., metallothionein IIA promoter; heat shock promoter, steroid/thyroid hormone/retinoic acid response elements) or from mammalian viruses (e.g., the adenovirus late promoter; mouse mammary tumor virus LTR) are used.
  • the invention includes in vivo methods.
  • a cell such as an undesirably proliferating cell or cell proliferative disorder to which an antibody or functional fragment binds, such as an antibody produced by a hybridoma set forth herein, or that includes any of SEQ ID NOs: 1 to 58, can be present in a subject, such as a mammal (e.g., a human subject).
  • a subject having such cells can therefore be treated by administering, for example, an antibody, or subsequence or fragment thereof, that binds to such cells.
  • a method includes administering to a subject an amount of an antibody, effective to treat the undesirable cell proliferation or a cell proliferative or cell hyperproliferative disorder in the subject.
  • Methods include combining the antibodies set forth herein with each other or a second therapeutic agent.
  • cell proliferative disorder and “cellular hyperproliferative disorder” and grammatical variations thereof, when used in reference to a cell, tissue or organ, refers to any undesirable, excessive or abnormal cell, tissue or organ growth, proliferation, differentiation or survival.
  • a hyperproliferative cell denotes a cell whose growth, proliferation, or survival is greater than desired, such as a reference normal cell, e.g., a cell that is of the same tissue or organ but is not a hyperproliferative cell, or a cell that fails to differentiate normally.
  • Undesirable cell proliferation and hyperproliferative disorders include diseases and physiological conditions, both benign hyperplastic conditions characterized by undesirable, excessive or abnormal cell numbers, cell growth, cell proliferation, cell survival or differentiation in a subject.
  • Specific examples of such disorders include metastatic and non-metastatic neoplasia, tumors and cancers (malignancies).
  • a method includes administering to a subject a an antibody, as represented by an antibody produced by a hybridoma, or heavy and light chain sequences set forth in any of SEQ ID NOs: 1 to 58, in an amount effective to treat the cell proliferative or cellular hyperproliferative disorder in the subject.
  • the disorder is a neoplasia, tumor or metastatic or non-metastatic cancer (malignancy).
  • the disorder affects or is present in part at least in breast, lung, thyroid, head and neck, nasopharynx, nose or sinuses, brain, spine, adrenal gland, thyroid, lymph, gastrointestinal (mouth, esophagus, stomach, duodenum, ileum, jejunum (small intestine), colon, rectum), genito-urinary tract (uterus, ovary, cervix, bladder, testicle, penis, prostate), kidney, pancreas, adrenal gland, liver, bone, bone marrow, lymph, blood, muscle, skin, or the hematopoetic system.
  • tumor refers to a cell or population of cells whose growth, proliferation or survival is greater than growth, proliferation or survival of a normal counterpart cell, e.g. a cell proliferative or differentiative disorder. Typically, the growth is uncontrolled.
  • malignancy refers to invasion of nearby tissue.
  • metastasis refers to spread or dissemination of a tumor, cancer or neoplasia to other sites, locations or regions within the subject, in which the sites, locations or regions are distinct from the primary tumor or cancer.
  • invention methods can be used to reduce or inhibit metastasis of a primary tumor or cancer to other sites, or the formation or establishment of metastatic tumors or cancers at other sites distal fiom the primary tumor or cancer thereby inhibiting or reducing tumor or cancer relapse or tumor or cancer progression.
  • methods of the invention include, amoung other things, 1) reducing or inhibiting growth, proliferation, mobility or invasiveness of tumor or cancer cells that potentially or do develop metastases (e.g, disseminated tumor cells, DTC); 2) reducing or inhibiting formation or establishment of metastases arising from a primary tumor or cancel to one or more other sites, locations or regions distinct from the primary tumor or cancer; 3) reducing or inhibiting growth or proliferation of a metastasis at one or more other sites, locations or regions distinct from the primary tumor or cancer after a metastasis has formed or has been established; and 4) reducing or inhibiting formation or establishment of additional metastasis after the metastasis has been formed or established
  • Neoplasias, tumors and cancers include a sarcoma, carcinoma, adenocaicinoma, melanoma, myeloma, blastoma, glioma, lymphoma or leukemia.
  • Exemplary cancers include, for example, carcinoma, sarcoma, adenocarcinoma, melanoma, neural (blastoma, glioma), mesothelioma and reticuloendothelial, lymphatic or haematopoietic neoplastic disorders (e.g., myeloma, lymphoma or leukemia).
  • a neoplasm, tumor or cancer includes a lung adenocarcinoma, lung carcinoma, diffuse or interstitial gastric carcinoma, colon adenocarcinoma, prostate adenocarcinoma, esophagus carcinoma, breast carcinoma, pancreas adenocarcinoma, ovarian adenocarcinoma, or uterine adenocarcinoma.
  • Neoplasia, tumors and cancers include benign, malignant, metastatic and non-metastatic types, and include any stage (I, II, III, IV or V) or grade (Gl, G2, G3, etc.) of neoplasia, tumor, or cancer, or a neoplasia, tumor, cancer or metastasis that is progressing, worsening, stabilized or in remission.
  • Neoplasias, tumors and cancers can arise from a multitude of primary tumor types, including but not limited to breast, lung, thyroid, head and neck, nasopharynx, nose or sinuses, brain, spine, adrenal gland, thyroid, lymph, gastrointestinal (mouth, esophagus, stomach, duodenum, ileum, jejunum (small intestine), colon, rectum), gemto-urinary tract (uterus, ovary, cervix, bladder, testicle, penis, prostate), kidney, pancreas, adrenal gland, liver, bone, bone marrow, lymph, blood, muscle, skin, and the hematopoetic system, and may metastasize to secondary sites.
  • primary tumor types including but not limited to breast, lung, thyroid, head and neck, nasopharynx, nose or sinuses, brain, spine, adrenal gland, thyroid, lymph, gastrointestinal (mouth, esophagus, stomach, duodenum, ileum, jejunum (small intestin
  • a "solid neoplasia, tumor or cancer” refers to neoplasia, tumor or cancer (e.g., metastasis) that typically aggregates together and forms a mass.
  • specific examples include visceral tumors such as melanomas, breast, pancreatic, uterine and ovarian cancers, testicular cancer, including seminomas, gastric or colon cancer, hepatomas, adrenal, renal and bladder carcinomas, lung, head and neck cancers and brain tumors/cancers.
  • Carcinomas refer to malignancies of epithelial or endocrine tissue, and include respiratory system carcinomas, gastrointestinal system carcinomas, genitourinary system carcinomas, testicular carcinomas, breast carcinomas, prostatic carcinomas, endocrine system carcinomas, and melanomas.
  • the term also includes carcinosarcomas, e.g., which include malignant tumors composed of carcinomatous and sarcomatous tissues.
  • Adenocarcinoma includes a carcinoma of a glandular tissue, or in which the tumor forms a gland like structure.
  • Melanoma refers to malignant tumors of melanocytes and other cells derived from pigment cell origin that may arise in the skin, the eye (including retina), or other regions of the body. Additional carcinomas can form from the uterine/cervix, lung, head/neck, colon, pancreas, testes, adrenal gland, kidney, esophagus, stomach, liver and ovary.
  • Sarcomas refer to malignant tumors of mesenchymal cell origin.
  • exemplary sarcomas include for example, lymphosarcoma, liposarcoma, osteosarcoma, chondrosarcoma, leiomyosarcoma, rhabdomyosarcoma and fibrosarcoma.
  • Neural neoplasias include glioma, glioblastoma, meningioma, neuroblastoma, retinoblastoma, astrocytoma, oligodendrocytoma
  • neoplasias, tumors and cancers amenable to treatment include malignant and non-malignant neoplasias, tumors and cancers, and metastasis.
  • adenocarcinoma or a squamous cell carcinoma such as a stomach adenocarcinoma, a lung adenocarcinoma, a pancreas adenocarcinoma, a colon adenocarcinoma, a breast adenocarcinoma, or an esophagus squamous cell carcinoma.
  • a "liquid neoplasia, tumor or cancer” refers to a neoplasia, tumor or cancer of the reticuloendothelial or hematopoetic system, such as a lymphoma, myeloma, or leukemia, or a neoplasia that is diffuse in nature.
  • leukemias include acute and chronic lymphoblastic, myeolblastic and multiple myeloma.
  • diseases arise from poorly differentiated acute leukemias, e.g., erythroblastic leukemia and acute megakaryoblastic leukemia.
  • lymphoid malignancies include, but are not limited to, acute lymphoblastic leukemia (APML), acute myelogenous leukemia (AML) and chronic myelogenous leukemia (CML); lymphoid malignancies include, but are not limited to, acute lymphoblastic leukemia (ALL), which includes B-lineage ALL and T-lineage ALL, chronic lymphocytic leukemia (CLL), prolymphocytic leukemia (PLL), hairy cell leukemia (HLL) and Waldenstrom's macroglobulinemia (WM).
  • ALL acute lymphoblastic leukemia
  • ALL which includes B-lineage ALL and T-lineage ALL
  • CLL chronic lymphocytic leukemia
  • PLL prolymphocytic leukemia
  • HLL hairy cell leukemia
  • W Waldenstrom's macroglobulinemia
  • Specific malignant lymphomas include, non-Hodgkin lymphoma and variants, peripheral T cell lymphomas, adult T cell leukemia/lymphoma (ATL), cutaneous T-cell lymphoma (CTCL), large granular lymphocytic leukemia (LGF), Hodgkin's disease and Reed-Sternberg disease.
  • the te ⁇ ns "treat,” “treating,” “treatment” and grammatical variations thereof mean subjecting an individual patient to a protocol, regimen, process or remedy, in which it is desired to obtain a physiologic response or outcome in that patient. Since every treated patient may not respond to a particular treatment protocol, regimen, process or remedy, treating does not require that the desired physiologic response or outcome be achieved in each and every patient or patient population. Accordingly, a given patient or patient population may fail to respond or respond inadequately to treatment.
  • Methods of the invention may be practiced by any mode of administration or by any route, systemic, regional and local administration.
  • Exemplary administration routes include intravenous, intrarterial, intradermal, intramuscular, subcutaneous, intra-pleural, transdermal (topical), transmucosal, intra-cranial, intra-spinal, intra-ocular, rectal, oral (alimentary) and mucosal.
  • Methods of the invention include, among other things, methods that provide a detectable or measurable improvement in a condition of a given subject, such as alleviating or ameliorating one or more adverse (physical) symptoms or consequences associated with the presence of a cell proliferative or cellular hyperprohferative disorder, neoplasm, tumor or cancer, or metastasis, i.e., a therapeutic benefit or a beneficial effect.
  • a therapeutic benefit or beneficial effect is any objective or subjective, transient, temporary, or long-term improvement in the condition or pathology, or a reduction in onset, severity, duration or frequency of an adverse symptom associated with or caused by cell proliferation or a cellular hyperprohferative disorder such as a neoplasm, tumor or cancer, or metastasis.
  • a satisfactory clinical endpomt of a treatment method in accordance with the invention is achieved, for example, when there is an incremental or a partial reduction in severity, duration or frequency of one or more associated pathologies, adverse symptoms or complications, or inhibition or reversal of one or more of the physiological, biochemical or cellular manifestations or characteristics of cell proliferation or a cellular hyperproliferative disorder such as a neoplasia, tumor or cancer, or metastasis.
  • a therapeutic benefit or improvement therefore be a cure, such as destruction of target proliferating cells (e.g., neoplasm, tumor or cancer, or metastasis) or ablation of one or more, most or all pathologies, adverse symptoms or complications associated with or caused by cell proliferation or the cellular hyperproliferative disorder such as a neoplasia, tumor or cancer, or metastasis.
  • target proliferating cells e.g., neoplasm, tumor or cancer, or metastasis
  • ablation of one or more, most or all pathologies, adverse symptoms or complications associated with or caused by cell proliferation or the cellular hyperproliferative disorder such as a neoplasia, tumor or cancer, or metastasis.
  • a therapeutic benefit or improvement need not be a cure or complete destruction of all target proliferating cells (e.g., neoplasia, tumor or cancer, or metastasis) or ablation of all pathologies, adverse symptoms or complications associated with or caused by cell proliferation or the cellular hyperprohferative disorder such as a neoplasia, tumor or cancer, or metastasis
  • target proliferating cells e.g., neoplasia, tumor or cancer, or metastasis
  • ablation of all pathologies, adverse symptoms or complications associated with or caused by cell proliferation or the cellular hyperprohferative disorder such as a neoplasia, tumor or cancer, or metastasis
  • Foi example partial destruction of a tumor or cancer cell mass, or a stabilization of the tumor or cancer mass, size or cell numbers by inhibiting progression or worsening of the tumor or cancer, can reduce mortality and prolong lifespan even if only for a few days, weeks or months, even though a portion or the
  • therapeutic benefit include a reduction in neoplasia, tumor or cancer, or metastasis volume (size or cell mass) or numbers of cells, inhibiting or preventing an increase in neoplasia, tumor or cancer volume (e.g., stabilizing), slowing or inhibiting neoplasia, tumor or cancer progression, worsening or metastasis, stimulating, inducing or increasing neoplasia, tumor or cancer cell lysis or apoptosis or inhibiting neoplasia, tumor or cancer proliferation, growth or metastasis.
  • An invention method may not take effect immediately.
  • treatment may be followed by an increase in the neoplasia, tumor or cancer cell numbers or mass, but over time eventual stabilization or reduction in tumor cell mass, size or numbers of cells in a given subject may subsequently occur after cell lysis or apoptosis of the neoplasia, tumor or cancer, or metastasis.
  • Additional adverse symptoms and complications associated with neoplasia, tumor, cancer and metastasis that can be inhibited, reduced, decreased, delayed or prevented include, for example, nausea, lack of appetite, lethargy, pain and discomfort.
  • a partial or complete decrease or reduction in the severity, duration or frequency of an adverse symptom or complication associated with or caused by a cellular hyperproliferative disorder, an improvement in the subjects well being, such as increased energy, appetite, psychological well being, are all particular non-limiting examples of therapeutic benefit.
  • a therapeutic benefit or improvement therefore can also include a subjective improvement in the quality of life of a treated subject.
  • a method reduces or decreases neoplasia, tumor or cancer, or metastasis volume, inhibits or prevents an increase in neoplasia, tumor or cancer volume, inhibits or delays neoplasia, tumor or cancer progression or worsening, stimulates neoplasia, tumor or cancer, or metastasis cell lysis or apoptosis, or inhibits, reduces, decreases or delays neoplasia, tumor or cancer proliferation or metastasis.
  • a method prolongs or extends lifespan of the subject.
  • a method improves the quality of life of the subject.
  • a biopsied sample containing a neoplasia, tumor or cancer, or metastasis e.g., blood or tissue sample
  • a biopsied sample containing a neoplasia, tumor or cancer, or metastasis can establish neoplastic, tumor or cancer cell volume or cell numbers, and therefore whether a reduction or stabilization in mass or numbers of neoplastic, tumor or cancer cells or inhibition of neoplasia, tumor or cancer cell proliferation, growth or survival (apoptosis) has occurred.
  • invasive and non- invasive imaging methods can ascertain neoplasia, tumor or cancer size or volume.
  • Examination of blood or serum, for example, for populations, numbers and types of cells can establish whether a reduction or stabilization in mass or numbers of neoplastic, tumor or cancer cells or inhibition of neoplastic, tumor or cancer proliferation, growth or survival (apoptosis) has occurred.
  • Antibodies and functional fragments can be combined with any other composition or combined with any other method other treatment or therapy that provides a desired effect.
  • treatments and therapies that have been characterized as having an anti-cell proliferative activity or function are applicable.
  • exemplary treatments and therapies include anti-cell proliferative or immune enhancing agents or drugs.
  • the methods of treatment and therapy can be performed prior to, substantially contemporaneously with any other method, for example, an anti- cell proliferative or anti-cellular hyperproliferative disorder (e.g., a neoplasia, tumor or cancer, or metastasis).
  • an anti- cell proliferative or anti-cellular hyperproliferative disorder e.g., a neoplasia, tumor or cancer, or metastasis.
  • antibodies and functional fragments can be administered prior to, substantially contemporaneously with any other composition or method.
  • the invention therefore provides combination compositions (e.g., two or more antibodies or functional fragments as set forth herein, an antibody or functional fragment as set forth herein and a second therapeutic agent, etc.) and combination methods, in which any of the antibodies, functional fragments, and modified and variant forms, are used in a combination with any therapeutic agent or composition, therapeutic regimen, treatment protocol, such as an anti-cell proliferative agent, drug or protocol, set forth herein or known in the art.
  • any therapeutic agent or composition, therapeutic regimen, treatment protocol such as an anti-cell proliferative agent, drug or protocol, set forth herein or known in the art.
  • a method includes administering an antibody or functional fragment, and an anti-cell proliferative or immune enhancing treatment, agent or drug.
  • the anti-cell proliferative or immune enhancing treatment, agent or drug can be administered prior to, substantially contemporaneously with or following administration of an antibody or functional fragment.
  • an "anti-cell proliferative,” “anti-neoplastic,” “anti-tumor,” or “anti-cancer” treatment, therapy, activity or effect means any therapy, treatment regimen, agent, drug, protocol or process that is useful in treating pathologies, adverse symptoms or complications associated with or caused by abnormal or undesirable cell proliferation (cell hyperproliferation), a cellular hyperproliferative disorder, neoplasia, tumor or cancer, or metastasis.
  • Particular therapies, treatment regimens, agents, drugs, protocol or processes can inhibit, decrease, slow, reduce, delay, or prevent cell proliferation, cell growth, cellular hyperproliferation, neoplastic, tumor, or cancer (malignant) growth, proliferation, survival or metastasis.
  • Such treatments, therapies, regimens, protocols, agents and drugs can operate by disrupting, reducing, inhibiting or delaying cell cycle progression or cell proliferation or growth: increasing, stimulating or enhancing cell apoptosis, lysis or death; inhibiting nucleic acid or protein synthesis or metabolism; reducing, decreasing, inhibiting or delaying cell division; or decreasing, reducing or inhibiting cell survival, or production or utilization of a cell survival factor, growth factor or signaling pathway (extracellular or intracellular).
  • anti-cell proliferative treatments and therapies include chemotherapy, immunotherapy, radiotherapy (ionizing or chemical), local or regional thermal (hyperthermia) therapy and surgical resection.
  • anti-cell proliferative agents and drugs include alkylating agents, anti-metabolites, plant extracts, plant alkaloids, nitrosoureas, hormones (steroids), nucleoside and nucleotide analogues.
  • microbial toxins include bacterial cholera toxin, pertussis toxin, anthrax toxin, diphtheria toxin, and plant toxin ricin.
  • drugs include cyclophosphamide, azathioprine, cyclosporin A, melphalan, chlorambucil, mechlorethamine, busulphan, methotrexate, 6- mercaptopurine, thioguanine, 5-fluorouracil (5-FU), 5-fluorouridine, cytosine arabinoside, AZT, 5-azacytidine (5-AZC) and 5-azacytidine related compounds, bleomycin, actinomycin D, mithramycin, mitomycin C, carmustine, calicheamicin, lomustine, semustine, streptozotocin, teniposide, etoposide, hydroxyurea, cisplatin, carboplatin, levamisole, mitotane, procarbazine, dacarbazine, taxol, vinblastine, vincristine, vindesine, doxorubicin, daunomycin and dibromomanni
  • Radiotherapy includes internal or external delivery to a subject.
  • alpha, beta, gamma and X-rays can administered to the subject externally without the subject internalizing or otherwise physically contacting the radioisotope.
  • Specific examples of X-ray dosages range from daily doses of 50 to 200 roentgens for prolonged periods of time (3 to 5/week), to single doses of 2000 to 6000 roentgens.
  • Radionuclides include, for example, 47 Sc 67 Cu, 72 Se, 88 Y 5 90 Sr, 90 Y, 97 Ru, 99 Tc, 105 Rh, " 1 In, 125 I, 131 I, 149 Tb, 153 Sm, 186 Re, 188 Re, 194 Os, 203 Pb, 21 1 At, 212 Bi, 213 Bi, 212 Pb, 223 Ra 5 225 Ac, 227 Ac, and 228 Th.
  • Antibodies that bind to tumor cells are a particular example of an anti-cell proliferative treatment or therapy.
  • Anti-tumor antibodies include, for example, M195 antibody which binds to leukemia cell CD33 antigen (U.S. Patent No. 6,599,505); monoclonal antibody DS6 which binds to ovarian carcinoma CA6 tumor-associated antigen (U.S. Patent No. 6,596,503); human IBD 12 monoclonal antibody which binds to epithelial cell surface H antigen (U.S. Patent No. 4,814,275); and BR96 antibody which binds to Le ⁇ carbohydrate epitope expressed by colon, breast, ovary, and lung carcinomas.
  • anti-tumor antibodies that can be employed include, for example, Herceptin (anti-Her-2 neu antibody), Rituxan®, Zevalin, Bevacizumab (Avastin), Bexxar, Campath®, Oncol ym, 17- IA (Edrecolomab), 3F8 (anti-neuroblastoma antibody), MDX- CTLA4, IMC-C225 (Cetuximab) and Mylotarg.
  • the term "immune enhancing,” when used in reference to a treatment, therapy, agent or drug means that the treatment, therapy, agent or drug provides an increase, stimulation, induction or promotion of an immune response, humoral or cell-mediated.
  • Such therapies can enhance immune response generally, or enhance immune response to a specific target, e.g., a cell proliferative or cellular hyperproliferative disorder such as a neoplasia, tumor or cancer, or metastasis.
  • immune enhancing agents include antibody, cell growth factors, cell survival factors, cell differentiative factors, cytokines and chemokines. Additional examples of immune enhancing agents and treatments include immune cells such as lymphocytes, plasma cells, macrophages, dendritic cells, NK cells and B-cells that either express antibody against the cell proliferative disorder or otherwise are likely to mount an immune response against the cell proliferative disorder.
  • Cytokines that enhance or stimulate immunogenicity include IL-2, IL- l ⁇ , IL- l ⁇ , IL-3, IL-6, IL-7, granulocyte-macrophage-colony stimulating factor (GMCSF), IFN- ⁇ , IL- 12, TNF- ⁇ , and TNF ⁇ , which are also non-limiting examples of immune enhancing agents.
  • Chemokines including MIP-loc, MIP-I ⁇ , RANTES, SDF-I, MCP-L MCP-2, MCP-3, MCP-4, eotaxin, eotaxin-2, 1-3O9/TCA3, ATAC, HCC-I, HCC-2, HCC- 3, PARC, TARC, LARC/MIP-3 ⁇ , CK ⁇ , CK ⁇ 6, CK ⁇ 7, CK ⁇ 8, CK ⁇ 9, CK ⁇ l 1, CK ⁇ l2, ClO, IL-8, ENA-78, GROoc, GRO ⁇ , GCP-2, PBP/CTAPIII ⁇ -TG/NAP-2, Mig, PBSF/SDF-L and lymphotactin are further non-limiting examples of immune enhancing agents.
  • Methods of the invention also include, among other things, methods that result in a reduced need or use of another treatment protocol or therapeutic regimen, process or remedy.
  • a method of the invention has a therapeutic benefit if in a given subject it results in a less frequent or reduced dose or elimination of an anti- cell proliferative (e.g., anti-neoplastic, anti-tumor or anti-cancer) or immune enhancing treatment or therapy, such as a chemotherapeutic drug, radiotherapy, immunotherapy, or surgery for neoplasia, tumor or cancer, or metastasis treatment or therapy.
  • an anti- cell proliferative e.g., anti-neoplastic, anti-tumor or anti-cancer
  • immune enhancing treatment or therapy such as a chemotherapeutic drug, radiotherapy, immunotherapy, or surgery for neoplasia, tumor or cancer, or metastasis treatment or therapy.
  • an anti-cell proliferative e.g., anti-neoplastic, anti-tumor, anti-cancer or anti-metastasis
  • a method includes administering to a subject an antibody, as represented by an antibody produced by a hybridoma, or heavy and light chain sequences set forth in any of SEQ ID NOs: 1 to 58, in an amount effective to treat a cellular hyperproliferative disorder (e.g., a neoplasia, tumor or cancer, or metastasis), and to reduce or eliminate need for an anti-cell proliferative (anti-neoplasia, anti-tumor or anticancer, or anti-metastasis) or immune-enhancing therapy.
  • the methods can be performed prior to, substantially contemporaneously with or following administration of an anti-neoplastic, tumor, cancer or metastasis, or immune- enhancing therapy.
  • the doses or "amount effective" or “amount sufficient” in a method of treatment or therapy in which it is desired to achieve a therapeutic benefit or improvement includes, for example, any objective or subjective alleviation or amelioration of one, several or all pathologies, adverse symptoms or complications associated with or caused by the target (e.g., cellular hyperproliferative disorder), to a measurable or detectable extent, although preventing, inhibiting or delaying a progression or worsening of the target (e.g., cellular hyperproliferative disorder) pathology, adverse symptom or complication, is a satisfactory outcome.
  • the target e.g., cellular hyperproliferative disorder
  • the amount in the case of a cellular hyperproliferative disorder, the amount will be sufficient to provide a therapeutic benefit to a given subject or to alleviate or ameliorate a pathology, adverse symptom or complication of the disorder in a given subject.
  • the dose may be proportionally increased or reduced as indicated by the status of treatment or therapeutic target (e.g., cellular hyperproliferative disorder) or any side effect(s) of the treatment or therapy.
  • Exemplary non-limiting amounts are in a range of about 0.1 mg/kg to about 100 mg/kg, and any numerical value or range or value within such ranges. Greater or lesser amounts (doses) can be administered, for example, 0.01-500 mg/kg, and any numerical value or range or value within such ranges. Additional exemplary non-limiting amounts (doses) range from about 0.5- 50 mg/kg, 1.0-25 mg/kg, 1.0-10 mg/kg, and any numerical value or range or value within such ranges.
  • Methods of the invention may be practiced one or more times (e.g., 1-10, 1-5 or 1-3 times) per day, week, month, or year.
  • times e.g., 1-10, 1-5 or 1-3 times
  • An exemplary non-limiting dosage schedule is 1-7 times per week, for 1, 23, 4, 5, 6, 7, 8, 9, 10, 15, 20 or more weeks, and any numerical value or range or value within such ranges.
  • Amounts effective or sufficient will therefore depend at least in part upon the disorder treated (e.g., cell proliferation, benign hyperplasia or a neoplasia, tumor or cancer and the type or stage, e.g., the tumor or cancer grade and if it is advanced, late or early stage), the therapeutic effect desired, as well as the individual subject (e.g., the bioavailability within the subject, gender, age, etc.) and the subject's response to the treatment based upon genetic and epigenetic variability (e.g., pharmacogenomics).
  • the disorder treated e.g., cell proliferation, benign hyperplasia or a neoplasia, tumor or cancer and the type or stage, e.g., the tumor or cancer grade and if it is advanced, late or early stage
  • the therapeutic effect desired e.g., the individual subject (e.g., the bioavailability within the subject, gender, age, etc.) and the subject's response to the treatment based upon genetic and epigenetic variability (
  • Cell toxicity and viability can be measured in a variety of ways on the basis of colorimetric, luminescent, radiometric, or fluorometric assays known in the art.
  • Colori ⁇ ietric techniques for determining cell viability include, for example. Trypan Blue exclusion. In brief, cells are stained with Trypan Blue and counted using a hemocytometer. Viable cells exclude the dye whereas dead and dying cells take up the blue dye and are easily distinguished under a light microscope. Neutral Red is adsorbed by viable cells and concentrates in cell lysosomes; viable cells can be determined with a light microscope by quantitating numbers of Neutral Red stained cells.
  • Fluorometric techniques for determining cell viability include, for example, propidium iodide, a fluorescent DNA intercalating agent. Propidium iodide is excluded from viable cells but stains the nucleus of dead cells. Flow cytometry of propidium iodide labeled cells can then be used to quantitate viable and dead cells. Release of lactate dehydrogenase (LDH) indicates structural damage and death of cells, and can be measured by a spectrophotometric enzyme assay. Bromodeoxyuridine (BrdU) is incorporated into newly synthesized DNA and can be detected with a fluorochrome-labeled antibody.
  • LDH lactate dehydrogenase
  • the fluorescent dye Hoechst 33258 labels DNA and can be used to quantitate proliferation of cells (e.g., flow cytometry). Quantitative incorporation of the fluorescent dye carboxyfluorcscein diacetate succinimidyl ester (CFSE or CFDA-SE) can provide cell division analysis (e.g., flow cytometry). This technique can be used either in vitro or in vivo. 7-aminoactinomycin D (7-AAD) is a fluorescent intercalator that undergoes a spectral shift upon association with DNA, and can provide cell division analysis (e.g., flow cytometry).
  • 7-AAD 7-aminoactinomycin D
  • Radiometric techniques for determining cell proliferation include, for example, [ 3 H]-Thymidine, which is incorporated into newly synthesized DNA of living cells and frequently used to determine proliferation of cells. Chromium ( 31 Cr)-release from dead cells can be quantitated by scintillation counting in order to quantitate cell viability.
  • Luminescent techniques for determining cell viability include, for example, the CellTiter-Glo luminescent cell viability assay (Promega Madison WI). This technique quantifies the amount of ATP present to determine the number of viable cells.
  • kits for determining cell viability and cell proliferation include, for example, Cell Proliferation Biotrak ELISA (Amersham Biosciences Piscataway, NJ); the Guava ViaCountTM Assay, which provides rapid cell counts and viability determination based on differential uptake of fluorescent reagents (Guava Technologies, Hayward, CA); the CyQU ANT® Cell Proliferation Assay Kit (Molecular Probes. Inc., Eugene, OR); and the CytoLux Assay Kit (PerkinElmer Life Sciences Inc., Boston, MA).
  • the DELFIA® Assay Kits can determine cell proliferation and viability using a time-resolved fluorometric method.
  • the QuantosTM Cell Proliferation Assay is a fluorescence-based assay that measures the fluorescence of a DNA-dye complex from lysed cells (Stratagene, La Jolla, CA).
  • the CellTiter-Glo cell viability assay is a luminescent assay for measuring cell viability (Promega, Madison WI).
  • subject and patient are used interchangeably herein and refer to animals, typically mammals, such as humans, non-human primates (gorilla, chimpanzee, orangutan, macaque, gibbon), domestic animals (dog and cat), farm and ranch animals (horse, cow, goat, sheep, pig), laboratory and experimental animals (mouse, rat, rabbit, guinea pig).
  • Subjects include disease model animals (e.g., such as mice, rats and non-human primates) for studying in vivo efficacy (e.g., a neoplasia, tumor or cancer, or metastasis animal model).
  • Human subjects include children, for example, newborns, infants, toddlers and teens, between the ages of 1 and 5, 5 and 10 and 10 and 18 years, adults between the ages of 18 and 60 years, and the elderly, for example, between the ages of 60 and 65, 65 and 70 and 70 and 100 years.
  • Subjects include mammals (e.g., humans) in need of treatment, that is, they have undesirable or aberrant cell proliferation (cell hyperproliferation) or a cellular hyperproliferative disorder. Subjects also include those at risk of having a undesirable cell proliferation or a cellular hyperproliferative disorder. Subjects further include a subject in need of an anti-cell proliferative or immune enhancing treatment or therapy due to a lab or clinical diagnosis warranting such treatment, subjects undergoing an anti cell proliferative or immune enhancing therapy, and subjects having undergone an anti-cell proliferative or immune enhancing therapy and are at risk of relapse oi recurrence
  • At risk subjects include those with a family history, genetic predisposition, or who have suffered a previous affliction with a cell proliferative or cellular hyperprohferative disorder (e g., a benign hyperplasia, neoplasm, tumor or cancer, or metastasis), and are at risk of relapse or recurrence.
  • a cell proliferative or cellular hyperprohferative disorder e g., a benign hyperplasia, neoplasm, tumor or cancer, or metastasis
  • At risk subjects further include environmental exposure to carcinogens or mutagens, such as smokers, or those in an occupational (industrial, chemical, agiicultural) setting
  • carcinogens or mutagens such as smokers, or those in an occupational (industrial, chemical, agiicultural) setting
  • Such subjects at risk for developing a cell proliferative or cellular hyperprohferative disorder such as neoplasia, tumor or cancer can be identified with genetic screens for tumor associated genes, gene deletions or gene mutations
  • Subjects that lack Brcal are at risk for developing breast cancer, for example.
  • Subjects at risk tor developing colon cancer have deleted or mutated tumor suppiessor genes, such as adenomatous polyposis cob (APC), for example
  • APC adenomatous polyposis cob
  • At risk subjects having particular genetic predisposition towards cell proliferative disorders are known (see, e g., The Genetic Basis of Human Cancer 2 nd ed by Bert Vogelstein (Editor), Kenneth W. Kinzler (Editor) (2002) McGraw-Hill Professional; The Molecular Basis of Human Cancer. Edited by WB Coleman and GJ Tsongalis (2001) Humana Press; and The Molecular Basis of Cancer. Mendelsohn et al , WB Saunders (1995))
  • At iisk subjects can therefore be tieated in order to inhibit or reduce the likelihood of developing a cell proliferative or cellular hyperprohferative disorder, or after havmg been cured of a cell proliferative disorder, suffering a relapse or recurrence of the same or a different cell proliferative or cellular hyperprohferative disorder.
  • the result of such treatment can be to reduce the ⁇ sk ol developing a cell proliferative or cellular hyperproliferative disorder, or to prevent a cell proliferative or cellular hyperprohferative disorder, or a pathology, adverse symptom or complication thereof in the treated at risk subject
  • kits including antibodies, functional fragments, modified and variants forms, nucleic acids, agents, drugs and pharmaceutical formulations, packaged into suitable packaging material, optionally in combination with instructions for using the kit components, e g , instructions for performing a method of the invention.
  • a kit includes an antibody, as represented by an antibody produced by a hybridoma, or heavy and light chain sequences set forth in any of SEQ ID NOs: 1 to 58.
  • the instructions are for treating undesirable cell proliferation or hyperproliferation, or a cellular hyperproliferative disorder.
  • the instructions are for treating a neoplasia, tumor or cancer, or metastasis.
  • a kit in a further embodiment, includes an antibody, as represented by an antibody produced by a hybridoma, or heavy and light chain sequences set forth in any of SEQ ID NOs: 1 to 58, and instructions for treating undesirable cell proliferation or hyperproliferation, or a cellular hyperproliferative disorder, and an anti-cell proliferative or immune enhancing treatment, agent or drug.
  • a kit includes an anti-neoplastic, anti-cancer or anti-tumor agent.
  • a kit includes an article of manufacture, for example, an article of manufacture for delivering the antibody or nucleic acid, anti-cell proliferative or immune enhancing treatment, agent or drug into a subject locally, regionally or systemically.
  • the term "packaging material” refers to a physical structure housing the components of the kit.
  • the packaging material can maintain the components sterilely, and can be made of material commonly used for such purposes (e.g., paper, corrugated fiber, glass, plastic, foil, ampules, etc.).
  • the label or packaging insert can include appropriate written instructions, for example, practicing a method of the invention, e.g., treating a cell proliferative or cellular hyperproliferative disorder, an assay for screening for, detecting or identifying an antigen (e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target.
  • an antigen e.g., CM-I target, CM-2 target, Pm-I target, Pm-2 target, Norm-1 target, Norm-2 target.
  • a cell or antigen e.g., CM-I target, CM-2 target,
  • a kit includes a label or packaging insert including instructions for practicing a method of the invention in solution, in vitro, in vivo, or ex vivo.
  • Instructions can therefore include instructions for practicing any of the methods of the invention described herein.
  • invention pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration to a subject to treat a cell proliferative or cellular hyperproliferative disorder, such as a neoplasia, tumor or cancer, or metastasis. Instructions may additionally include indications of a satisfactory clinical endpoint or any adverse symptoms or complications that may occur, storage information, expiration date, or any information required by regulatory agencies such as the Food and Drug Administration for use in a human subject.
  • the instructions may be on "printed matter," e.g., on paper or cardboard within the kit, on a label affixed to the kit or packaging material, or attached to a vial or tube containing a component of the kit.
  • Instructions may comprise voice or video tape and additionally be included on a computer readable medium, such as a disk (floppy diskette or hard disk), optical CD such as CD- or DVD-ROM/RAM, magnetic tape, electrical storage media such as RAM and ROM and hybrids of these such as magnetic/optical storage media.
  • kits can additionally include a buffering agent, a preservative, or a protein/nucleic acid stabilizing agent.
  • the kit can also include control components for assaying for activity, e.g., a control sample or a standard.
  • Each component of the kit can be enclosed within an individual container or in a mixture and all of the various containers can be within single or multiple packages.
  • Antibodies and functional fragments can be included in or employ pharmaceutical formulations. Such pharmaceutical formulations are useful for treatment of, or administration or delivery to, a subject in vivo or ex vivo.
  • compositions include “pharmaceutically acceptable” and “physiologically acceptable” carriers, diluents or excipients.
  • pharmaceutically acceptable and “physiologically acceptable” include solvents (aqueous or non-aqueous), solutions, emulsions, dispersion media, coatings, isotonic and absorption promoting or delaying agents, compatible with pharmaceutical administration.
  • Such formulations can be contained in a liquid; emulsion, suspension, syrup or elixir, or solid form; tablet (coated or uncoated), capsule (hard or soft), powder, granule, crystal, or microbead.
  • Supplementary compounds e.g., preservatives, antibacterial, antiviral and antifungal agents
  • compositions of the invention can be made to be compatible with a particular local, regional or systemic administration or delivery route.
  • pharmaceutical formulations include carriers, diluents, or excipients suitable for administration by particular routes.
  • routes of administration for compositions of the invention are parenteral, e.g., intravenous, intrarterial, intradermal, intramuscular, subcutaneous, intra-pleural. transdermal (topical), transmucosal, intra-cranial, intra-spinal, intra-ocular, rectal, oral (alimentary), mucosal administration, and any other formulation suitable for the treatment method or administration protocol.
  • Solutions or suspensions used for parenteral application can include: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents
  • antibacterial agents such as benzyl alcohol or methyl parabens
  • antioxidants such as ascorbic acid or sodium bisulfit
  • compositions for injection include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, NJ) or phosphate buffered saline (PBS).
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof.
  • Fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Antibacterial and antifungal agents include, for example, parabens, chlorobutanol, phenol, ascorbic acid and thimerosal.
  • Isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, sodium chloride can be included in the composition.
  • Including an agent which delays absorption, for example, aluminum monostcarate or gelatin can prolong absorption of injectable compositions.
  • Sterile injectable formulations can be prepared by incorporating the active composition in the required amount in an appropriate solvent with one or a combination of above ingredients.
  • dispersions are prepared by incorporating the active composition into a sterile vehicle containing a basic dispersion medium and any other ingredient.
  • methods of preparation include, for example, vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously prepared solution thereof.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sprays, inhalation devices (e.g., aspirators) or suppositories.
  • the active compounds are formulated into ointments, salves, gels, creams or patches.
  • the pharmaceutical formulations can be prepared with carriers that protect against rapid elimination from the body, such as a controlled release formulation or a time delay material such as glyceryl monostearate or glyceryl stearate.
  • a controlled release formulation or a time delay material such as glyceryl monostearate or glyceryl stearate.
  • the formulations can also be delivered using articles of manufacture such as implants and microencapsulated delivery systems to achieve local, regional or systemic delivery or controlled or sustained release.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations are known to those skilled in the art. The materials can also be obtained commercially from Alza Corporation (Palo Alto, CA). Liposomal suspensions (including liposomes targeted to cells or tissues using antibodies or viral coat proteins) can also be used as pharmaceutically acceptable carriers. These can be prepared according to known methods, for example, as described in U.S. Patent No. 4,522,811.
  • compositions used in accordance with the invention including proteins (antibodies), nucleic acid (inhibitory), treatments, therapies, agents, drugs and pharmaceutical formulations can be packaged in dosage unit form for ease of administration and uniformity of dosage.
  • dosage unit form refers to physically discrete units suited as unitary dosages treatment; each unit contains a quantity of the composition in association with the carrier, excipient, diluent, or vehicle calculated to produce the desired treatment or therapeutic (e.g., beneficial) effect.
  • the unit dosage forms will depend on a variety of factors including, but not necessarily limited to, the particular composition employed, the effect to be achieved, and the pharmacodynamics and pharmacogenomics of the subject to be treated.
  • contacting when used in reference to a composition such as a protein (e.g., antibody), material, sample, or treatment, means a direct or indirect interaction between the composition (e.g., protein such as an antibody) and the other referenced entity.
  • a particular example of direct interaction is binding.
  • a particular example of an indirect interaction is where the composition acts upon an intermediary molecule, which in turn acts upon the referenced entity.
  • contacting a cell e.g., that comprises a cellular hyperprol iterative disorder
  • an antibody includes allowing the antibody to bind to the cell, or allowing the antibody to act upon an intermediary (e.g., antigen) that in turn acts upon the cell.
  • testing and “measuring” and grammatical variations thereof are used interchangeably herein and refer to either qualitative or quantitative determinations, or both qualitative and quantitative determinations.
  • any means of assessing the relative amount, affinity or specificity of binding is contemplated, including the various methods set forth herein and known in the art. For example, antibody binding can be assayed or measured by an ELISA assay.
  • references to a range of 90-100% includes any numerical value or range within or encompassing such values, such as 91%, 92%, 93%, 94%, 95%, 95%, 97%, etc., as well as 91.1%, 91.2%, 91.3%, 91.4%, 91.5%, etc., 92.1%, 92.2%, 92.3%, 92.4%, 92.5%, etc., and any numerical range within such a range, such as 90-92%, 90-95%, 95-98%, 96-98%.
  • reference to a range of 1-5,000 fold includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, fold, etc., as well as 1.1, 1.2, 1.3, 1.4, 1.5, fold, etc., 2.1, 2.2, 2.3, 2.4, 2.5, fold, etc., and any numerical range within such a range, such as 1-2. 5-10, 10-50, 50-100, 100-500, 100-1000, 500-1000, 1000-2000, 1000-5000, etc.
  • reference to a range of KD 10 "5 M to about KD 10 "n M includes any numerical value or range within or encompassing such values.
  • the invention is generally disclosed herein using affirmative language to describe the numerous embodiments.
  • the invention also specifically includes embodiments in which particular subject matter is excluded, in full or in part, such as substances or materials, method steps and conditions, protocols, procedures, assays or analysis.
  • the invention is generally not expressed herein in terms of what the invention does not include, aspects that are not expressly included in the invention are nevertheless disclosed.
  • Tumor staining was determined for several human IgM antibodies on the human pancreatic carcinoma cell line BxPC-3. Conjugated antibodies at a final concentration of 400 ⁇ g/ml were directly given to 1 x 106 cells and incubated for 60 min on ice. Finally, the slides were mounted with Fluorescent Mounting Medium (DakoCytomation, Carpinteria, CA) and analyzed by confocal microscopy. Increased staining of BXPC-3 cells was observed with a combination of SAM-6 and Norm-2 and a combination of CM-I and Norm-2.
  • Trypan blue staining was used to determine antiproliferative effects of antibody combinations (so called cocktails) on the pancreatic tumor cell line BXPC-3. Trypsinized cells were diluted to 1 x 106 cells/ml in complete growth medium and 1 millilitre of this solution was given in to each well of a six well plate. Antibodies and controls (RPMI and Chrompure IgM) was given to the cells to an final volume of 3 ml. After 48h of incubation at 37°C in a humidified incubator attached cells were detached through brief exposure to trypsin/EDTA. Both floating and attached cells were pooled and counted. Only viable cells were counted using trypan blue. The activity of an antibody was defined as difference between the amount of cells in the control group and the amount of the antibody treated group in percentage. The data is illustrated in Figure 1. [02961 The data is summarized in Table 1 :
  • pancreatic cancer cells (BxPC-3) were pretreated with 5-FU for 24 h and then incubated with human antibodies. Trypsinized cells were diluted to 2x105 cells/ml in complete growth medium supplemented with 5 Fluorouracil (5-FU) in concentrations of 5 ⁇ g/ml, 2 ⁇ g/ml and l ⁇ g/ml. 50 ⁇ l of the cell suspension was added to each well of a 96-well plate.
  • 5-FU Fluorouracil
  • pancreatic cancer cells BxPC-3 and colon cancer cells (HT29) were pretreated with 5-FU for 24 h and then incubated with human antibodies. Trypsinized cells were diluted to 2x105 cells/ml in complete growth medium supplemented with 5 Fluorouracil (5-FU) in concentrations of 5 ⁇ g/ml, 2 ⁇ g/ml and l ⁇ g/ml. 50 ⁇ l of the cell suspension was added to each well of a 96-well plate.
  • 5-FU Fluorouracil
  • Cell lines BxPC-3 and HT29 were used for the analysis of NORM-2 and SAM-6 receptor expiession after pre-treatment with 5-FU. Cells were giown to subconfluency in complete medium, and then 5-Fluoruracil was added to a final concentration of 5 ⁇ g/ml. As a control, cells were incubated in RPMI-1640 medium with 10% FCS without 5-FU. Cells were harvested after 12 hours by detaching with Trypsin/EDTA and incubated on ice for Ih for recreation.
  • the cells were subsequently incubated on ice with NORM-2 or SAM-6 (both 200 ⁇ g/ml), and human unrelated IgM control antibody (Chrompure human IgM; Dianova, Hamburg, Germany) for 15 minutes. This was followed by incubation with a FITC-labeled rabbit anti-human IgM antibody (Dianova) respectively, for 15 minutes on ice. Antibodies were optimally diluted in PBS containing 0.01% sodiumazide. Cells were analyzed by flow cytometry (FACScan; Becton Dickinson, San Jose, California) The data is illustrated in Figure 3.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Oncology (AREA)
  • Biomedical Technology (AREA)
  • Microbiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • High Energy & Nuclear Physics (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

L’invention concerne des combinaisons d’anticorps et de fragments fonctionnels, ainsi que des procédés d’utilisation des anticorps et des fragments fonctionnels en combinaison (par ex., chaque anticorps administré individuellement à un sujet séquentiellement ou administré à un sujet comme composition en combinaison). Les anticorps comprennent IgM et IgG. IgM comprend des combinaisons d’IgM (par ex., deux IgM ou plus). IgG comprend des combinaisons d’IgG (par ex., deux IgG ou plus). Des combinaisons comprennent des combinaisons d’IgM (par ex., deux IgM ou plus), des combinaisons d’IgG (par ex., deux IgG ou plus) et des combinaisons d’IgM et d’IgG. Les procédés comprennent l’administration de combinaisons d’IgM (par ex., deux IgM ou plus), de combinaisons d’IgG (par ex., deux IgG ou plus) et de combinaisons d’IgM et d’IgG à un sujet.
PCT/IB2009/005352 2008-02-19 2009-02-19 Combinaisons d’anticorps et leurs procédés de fabrication et d’utilisation WO2009104100A2 (fr)

Applications Claiming Priority (20)

Application Number Priority Date Filing Date Title
US2986908P 2008-02-19 2008-02-19
US61/029,869 2008-02-19
US4395008P 2008-04-10 2008-04-10
US4396108P 2008-04-10 2008-04-10
US61/043,950 2008-04-10
US61/043,961 2008-04-10
US6188108P 2008-06-16 2008-06-16
US61/061,881 2008-06-16
US8481408P 2008-07-30 2008-07-30
US61/084,814 2008-07-30
US12/339,737 2008-12-19
US12/339,737 US20090202570A1 (en) 2007-12-21 2008-12-19 Pm-2 antibodies, functional fragments and methods for treating metastasis
US12/349,390 US20090258020A1 (en) 2008-01-07 2009-01-06 Antibody designated barb3, barb3 related antibodies, and methods of making and using same
US12/349,390 2009-01-06
US12/350,072 US20090291083A1 (en) 2008-01-07 2009-01-07 Barb4 target, antibody designated barb4, barb4 related antibodies, and methods of making and using same
US12/350,072 2009-01-07
US14335109P 2009-01-08 2009-01-08
US61/143,351 2009-01-08
US15114909P 2009-02-09 2009-02-09
US61/151,149 2009-02-09

Publications (2)

Publication Number Publication Date
WO2009104100A2 true WO2009104100A2 (fr) 2009-08-27
WO2009104100A3 WO2009104100A3 (fr) 2010-01-14

Family

ID=40985995

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2009/005352 WO2009104100A2 (fr) 2008-02-19 2009-02-19 Combinaisons d’anticorps et leurs procédés de fabrication et d’utilisation

Country Status (1)

Country Link
WO (1) WO2009104100A2 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010004438A3 (fr) * 2008-06-16 2010-03-11 Patrys Limited Anticorps anti-lm, fragments fonctionnels, antigène cible lm-1, et procédés pour les préparer et les utiliser
WO2010088739A1 (fr) * 2009-02-09 2010-08-12 Patrys Limited Variants de sam-6, cible et procédés d'utilisation correspondants
EP2374820A3 (fr) * 2003-11-14 2012-01-11 Patrys Limited Anticorps monoclonal humain reducteur de graisse
CN110187052A (zh) * 2019-03-08 2019-08-30 合肥学院 一种基于云计算的植物生长因素对其生长影响的分析方法
WO2021231237A3 (fr) * 2020-05-11 2021-12-16 Augmenta Bioworks, Inc. Anticorps anti sras-cov-2 et leurs utilisations

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0352500A2 (fr) * 1988-07-27 1990-01-31 Biotest Pharma Gmbh Procédé pour la production d une preparation à base d une immuno globuline polyclonale par voie intraveneuse avec un contenu éléve en IgM.
WO2004081027A2 (fr) * 2003-03-14 2004-09-23 Oncomab Gmbh Anticorps specifiques des neoplasmes et leurs utilisations
EP1531162A1 (fr) * 2003-11-14 2005-05-18 Heinz Vollmers Anticorps SAM-6, spécifique d'adénocarcinome, et ses utilisations
WO2005094159A2 (fr) * 2003-11-12 2005-10-13 Oncomab Gmbh Methodes d'identification d'anticorps specifiques de tumeurs et utilisations
US20070086999A1 (en) * 2003-05-16 2007-04-19 Gruskin Elliot A Compositions and methods including a recombinant human mab that promotes cns remyelination

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0352500A2 (fr) * 1988-07-27 1990-01-31 Biotest Pharma Gmbh Procédé pour la production d une preparation à base d une immuno globuline polyclonale par voie intraveneuse avec un contenu éléve en IgM.
US5190752A (en) * 1988-07-27 1993-03-02 Biotest Pharma Gmbh Intravenously administerable polyclonal immunoglobulin preparation containing igm and method of manufacture
WO2004081027A2 (fr) * 2003-03-14 2004-09-23 Oncomab Gmbh Anticorps specifiques des neoplasmes et leurs utilisations
US20070086999A1 (en) * 2003-05-16 2007-04-19 Gruskin Elliot A Compositions and methods including a recombinant human mab that promotes cns remyelination
WO2005094159A2 (fr) * 2003-11-12 2005-10-13 Oncomab Gmbh Methodes d'identification d'anticorps specifiques de tumeurs et utilisations
EP1531162A1 (fr) * 2003-11-14 2005-05-18 Heinz Vollmers Anticorps SAM-6, spécifique d'adénocarcinome, et ses utilisations

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
BRAENDLEIN S; ET AL: "HUMAN MONOCLONAL IGM ANTIBODIES WITH APOPTOTIC ACTIVITY ISOLATED FROM CANCER PATIENTS" HUMAN ANTIBODIES, IOS PRESS, AMSTERDAM, NL, vol. 11, no. 4, 1 January 2002 (2002-01-01), pages 107-119, XP009020163 ISSN: 1093-2607 *
BRAENDLEIN STEPHANIE; ET AL: "Natural IgM antibodies and immunosurveillance mechanisms against epithelial cancer cells in humans" CANCER RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, BALTIMORE, MD., US, [Online] vol. 63, no. 22, 15 November 2003 (2003-11-15), pages 7995-8005, XP002524676 ISSN: 0008-5472 internet Retrieved from the Internet: URL:http://cancerres.aacrjournals.org/cgi/content/full/63/22/7995> *
BRÄNDLEIN STEPHANIE; RAUSCHERT NICOLE; RASCHE LEO; DREYKLUFT ANGELA; HENSEL FRANK; CONZELMANN ERNST; MÜLLER-HERMELINK HANS-KONRAD;: "The human IgM antibody SAM-6 induces tumor-specific apoptosis with oxidized low-density lipoprotein." MOLECULAR CANCER THERAPEUTICS JAN 2007, vol. 6, no. 1, January 2007 (2007-01), pages 326-333, XP002545241 ISSN: 1535-7163 *
EMMANOUILIDES C; PEGRAM M; ROBINSON R; HECHT R; KABBINAVAR F; ISACOFF W: "Anti-VEGF antibody bevacizumab (Avastin) with 5FU/LV as third line treatment for colorectal cancer." TECHNIQUES IN COLOPROCTOLOGY NOV 2004, vol. 8 Suppl 1, November 2004 (2004-11), pages S50-S52, XP002545240 ISSN: 1123-6337 *
HURWITZ H I; ET AL: "BEVACIZUMAB IN COMBINATION WITH FLUOROURACIL AND LEUCOVORIN: AN ACTIVE REGIMEN FOR FIRST-LINE METASTATIC COLORECTAL CANCER" JOURNAL OF CLINICAL ONCOLOGY, AMERICAN SOCIETY OF CLINICAL ONCOLOGY, US, vol. 23, no. 15, 20 May 2005 (2005-05-20), pages 3502-3508, XP009050551 ISSN: 0732-183X *
TAKEDA KAZUYOSHI; OKUMURA KO; SMYTH MARK J: "Combination antibody-based cancer immunotherapy." CANCER SCIENCE SEP 2007, vol. 98, no. 9, September 2007 (2007-09), pages 1297-1302, XP002545239 ISSN: 1347-9032 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2374820A3 (fr) * 2003-11-14 2012-01-11 Patrys Limited Anticorps monoclonal humain reducteur de graisse
WO2010004438A3 (fr) * 2008-06-16 2010-03-11 Patrys Limited Anticorps anti-lm, fragments fonctionnels, antigène cible lm-1, et procédés pour les préparer et les utiliser
WO2010088739A1 (fr) * 2009-02-09 2010-08-12 Patrys Limited Variants de sam-6, cible et procédés d'utilisation correspondants
CN110187052A (zh) * 2019-03-08 2019-08-30 合肥学院 一种基于云计算的植物生长因素对其生长影响的分析方法
WO2021231237A3 (fr) * 2020-05-11 2021-12-16 Augmenta Bioworks, Inc. Anticorps anti sras-cov-2 et leurs utilisations
US11919944B2 (en) 2020-05-11 2024-03-05 Augmenta Biosciences, Inc. Antibodies for SARS-CoV-2 and uses thereof

Also Published As

Publication number Publication date
WO2009104100A3 (fr) 2010-01-14

Similar Documents

Publication Publication Date Title
ES2550757T3 (es) Anticuerpos que reconocen un epítopo que contiene carbohidratos en CD43 y ACE expresados en células cancerosas y métodos de uso de los mismos
KR102048718B1 (ko) 항-cxcr4 항체 및 항체-약물 접합체
RU2573897C2 (ru) Антитела к cxcr4 и их применение для лечения рака
CN110357961B (zh) 抗人4-1bb单克隆抗体及其制备方法和用途
BR112020022595A2 (pt) anticorpos específicos para gucy2c e usos dos mesmos
RU2595394C2 (ru) Гуманизированные антитела к cxcr4 для лечения рака
CN110467674B (zh) 抗ox40的全人抗体及其制备方法和用途
BRPI0306973B1 (pt) Anticorpos anti-igf-ir, seu método de produção e seus usos, hibridoma murino, ácido nucléico isolado, vetor, composição, kit ou conjunto, e método diagnóstico in vitro de doenças induzidas por uma superexpressão ou uma subexpressão do receptor igf-ir
PT1735348E (pt) Anticorpo anti-receptor do factor de crescimento humano
WO2009104100A2 (fr) Combinaisons d’anticorps et leurs procédés de fabrication et d’utilisation
CN113423736A (zh) 对muc18特异性的抗体
JP2020523414A (ja) Lgr5に結合する抗体薬物複合体
US9783599B2 (en) LM-antibodies, functional fragments, LM-1 target antigen, and methods for making and using same
US20090258020A1 (en) Antibody designated barb3, barb3 related antibodies, and methods of making and using same
WO2018047917A1 (fr) Effet antitumoral synergétique d'une combinaison de la protéine hmg, de l'anticorps anti-cd4 ou d'un agent de contrôle de point de contrôle immunitaire
JP7196311B2 (ja) 抗tim-3抗体とその利用
US20090202570A1 (en) Pm-2 antibodies, functional fragments and methods for treating metastasis
CN116942809A (zh) 一种抗her2抗体在制备治疗癌症的药物中的用途
US20090291083A1 (en) Barb4 target, antibody designated barb4, barb4 related antibodies, and methods of making and using same
CN115957321A (zh) 一种抗her2抗体在制备治疗癌症的药物中的用途

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09713351

Country of ref document: EP

Kind code of ref document: A2

122 Ep: pct application non-entry in european phase

Ref document number: 09713351

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE